US20060241123A1 - Substituted piperidine and piperazine derivatives as melanocortin-4 receptor modulators - Google Patents

Substituted piperidine and piperazine derivatives as melanocortin-4 receptor modulators Download PDF

Info

Publication number
US20060241123A1
US20060241123A1 US10/550,267 US55026704A US2006241123A1 US 20060241123 A1 US20060241123 A1 US 20060241123A1 US 55026704 A US55026704 A US 55026704A US 2006241123 A1 US2006241123 A1 US 2006241123A1
Authority
US
United States
Prior art keywords
alkyl
heterocyclyl
cycloalkyl
hydrogen
heteroaryl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/550,267
Inventor
Michael Soeberdt
Philipp Weyermann
Andreas Sprecher
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Santhera Pharmaceuticals Schweiz GmbH
Original Assignee
Santhera Pharmaceuticals Schweiz GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Santhera Pharmaceuticals Schweiz GmbH filed Critical Santhera Pharmaceuticals Schweiz GmbH
Assigned to SANTHERA PHARMACEUTICALS (SCHWEIZ) GMBH reassignment SANTHERA PHARMACEUTICALS (SCHWEIZ) GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SOEBERDT, MICHAEL, VON SPRECHER, ANDREAS, WEYERMANN, PHILIPP
Publication of US20060241123A1 publication Critical patent/US20060241123A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/10Spiro-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/438The ring being spiro-condensed with carbocyclic or heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/22Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4
    • C07D311/24Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings

Definitions

  • the present invention relates to novel substituted piperidine and piperazine derivatives as melanocortin-4 receptor modulators.
  • the compounds of the invention are either selective agonists or selective antagonists of the human melanocortin-4 receptor (MC-4R).
  • the agonists can be used for the treatment of disorders and diseases such as obesity, diabetes and sexual dysfunction, whereas the antagonists are useful for the treatment of disorders and diseases such as cancer cachexia, muscle wasting, anorexia, anxiety and depression.
  • MC-4R human melanocortin-4 receptor
  • MCs Melanocortins stem from pro-opiomelanocortin (POMC) via proteolytic cleavage.
  • POMC pro-opiomelanocortin
  • peptides range in size from 12 to 39 amino acids.
  • ⁇ -MSH acts as a neurotransmitter or neuromodulator in the brain.
  • MC peptides, particularly ⁇ -MSH have a wide range of effects on biological functions including feeding behavior, pigmentation and exocrine function.
  • MC-Rs melanocortin receptors
  • MC-1R was first found in melanocytes. Naturally occurring inactive variants of MC-1R in animals were shown to lead to alterations in pigmentation and a subsequent lighter coat color by controlling the conversion of phaeomelanin to eumelanin through the control of tyrosinase. From these and other studies, it is evident that MC-1R is an important regulator of melanin production and coat color in animals and skin color in humans.
  • the MC-2R is expressed in the adrenal gland representing the ACTH receptor.
  • the MC-2R is not a receptor for ⁇ -MSH but is the receptor for the adrenocorticotropic hormone I (ACTH I).
  • the MC-3R is expressed in the brain (predominately located in the hypothalamus) and peripheral tissues like gut and placenta, and knock-out studies have revealed that the MC-3R may be responsible for alterations in feeding behavior, body weight and thermogenesis.
  • the MC-4R is primarily expressed in the brain. Overwhelming data support the role of MC-4R in energy homeostasis. Genetic knock-outs and pharmacologic manipulation of MC-4R in animals have shown that agonizing the MC-4R causes weight loss and antagonizing the MC-4R produces weight gain (A. Kask, et al., “Selective antagonist for the melanocortin-4 receptor (HS014) increases food intake in free-feeding rats,” Biochem. Biophys. Res. Commun., 245: 90-93 (1998)).
  • MC-5R is ubiquitously expressed in many peripheral tissues including white fat, placenta and a low level of expression is also observed in the brain. However its expression is greatest in exocrine glands. Genetic knock-out of this receptor in mice results in altered regulation of exocrine gland function, leading to changes in water repulsion and thermoregulation. MC-5R knockout mice also reveal reduced sebaceous gland lipid production (Chen et al., Cell, 91: 789-798 (1997)).
  • MC-3R and MC-4R modulators have potent physiological effects besides their role in regulating pigmentation, feeding behavior and exocrine function.
  • ⁇ -MSH recently has been shown to induce a potent anti-inflammatory effect in both acute and chronic models of inflammation including inflammatory bowel-disease, renal ischemia/reperfusion injury and endotoxin-induced hepatitis.
  • Administration of ⁇ -MSH in these models results in substantial reduction of inflammation-mediated issue damage, a significant decrease in leukocyte infiltration and a dramatic reduction in elevated levels of cytokines and other mediators to near baseline levels.
  • ⁇ -MSH anti-inflammatory actions of ⁇ -MSH are mediated by MC-1R.
  • the mechanism by which agonism of MC-1R results in an anti-inflammatory response is likely through inhibition of the pro-inflammatory transcription activator, NF- ⁇ B.
  • NF- ⁇ B is a pivotal component of the pro-inflammatory cascade, and its activation is a central event in initiating many inflammatory diseases.
  • anti-inflammatory actions of ⁇ -MSH may be, in part, mediated by agonism of MC-3R and/or MC-5R.
  • MC-4R signaling is important in mediating feeding behavior (S. Q. Giraudo et al., “Feeding effects of hypothalamic injection of melanocortin-4 receptor ligands,” Brain Research, 80: 302-306 (1998)).
  • Further evidence for the involvement of MC-Rs in obesity includes: a) the agouti (A vy ) mouse which ectopically expresses an antagonist of the MC-1R.
  • MC-3R and MC-4R is obese, indicating that blocking the action of these three MC-R's can lead to hyperphagia and metabolic disorders; 2) MC-4R knockout mice (D.
  • MC-4R appears to play a role in other physiological functions as well, namely controlling grooming behavior, erection and blood pressure.
  • Erectile dysfunction denotes the medical condition of inability to achieve penile erection sufficient for successful intercourse.
  • the term “impotence” is often employed to describe this prevalent condition.
  • Synthetic melanocortin receptor agonists have been found to initiate erections in men with psychogenic erectile dysfunction (H. Wessells et al., “Synthetic Melanotropic Peptide Initiates Erections in Men With Psychogenic Erectile Dysfunction: Double-Blind, Placebo Controlled Crossover Study,” J. Urol., 160: 389-393, 1998).
  • Activation of melanocortin receptors of the brain appears to cause normal simulation of sexual arousal.
  • Evidence for the involvement of MC-R in male and/or female sexual dysfunction is detailed in WO/0074679.
  • Diabetes is a disease in which a mammal's ability to regulate glucose levels in the blood is impaired because the mammal has a reduced ability to convert glucose to glycogen for storage in muscle and liver cells. In Type I diabetes, this reduced ability to store glucose is caused by reduced insulin production.
  • Type II diabetes or “Non-Insulin Dependent Diabetes Mellitus” (NIDDM) is the form of diabetes which is due to a profound resistance to insulin stimulating or regulatory effect on glucose and lipid metabolism in the main insulin-sensitive tissues, muscle, liver and adipose tissue. This resistance to insulin responsiveness results in insufficient insulin activation of glucose uptake, oxidation and storage in muscle, and inadequate insulin repression of lipolysis in adipose tissue and of glucose production and secretion in liver.
  • NIDDM Non-Insulin Dependent Diabetes Mellitus
  • Hyperinsulemia is associated with hypertension and elevated body weight. Since insulin is involved in promoting the cellular uptake of glucose, amino acids and triglycerides from the blood by insulin sensitive cells, insulin insensitivity can result in elevated levels of triglycerides and LDL which are risk factors in cardiovascular diseases.
  • MC-4R agonists might be useful in the treatment of NIDDM and Syndrome X.
  • the MC-4 receptor is also of interest in terms of the relationship to stress and the regulation of emotional behavior, as based on the following findings. Stress initiates a complex cascade of responses that include endocrine, biochemical and behavioral events. Many of these responses are initiated by release of corticotropin-releasing factor (CRF) (Owen M J and Nemeroff C B (1991) Physiology and pharmacology of corticotrophin releasing factor. Pharmacol Rev 43:425-473).
  • CCF corticotropin-releasing factor
  • MCL0129 (1-[(S)-2-(4-Fluorophenyl)-2-(4-isopropylpiperadin-1-yl)ethyl]-4-[4-(2-methoxynaphthalen-1-yl)butyl]piperazine), a Novel and Potent Nonpeptide Antagonist of the Melanocortin-4 Receptor; Shigeyuki Chaki et al, J. Pharm. Exp. Ther. (2003)304(2), 818-26).
  • Chronic diseases such as malignant tumors or infections, are frequently associated with cachexia resulting from a combination of a decrease in appetite and a loss of lean body mass.
  • Extensive loss of lean body mass is often triggered by an inflammatory process and is usually associated with increased plasma levels of cytokines (e.g. TNF- ⁇ ), which increase the production of ⁇ -MSH in the brain.
  • cytokines e.g. TNF- ⁇
  • Activation of MC-4 receptors in the hypothalamus by ⁇ -MSH reduces appetite and increases energy expenditure.
  • Experimental evidence in tumor bearing mice suggests that cachexia can be prevented or reversed by genetic MC-4 receptor knockout or MC-4 receptor blockade.
  • the increased body weight in the treated mice is attributable to a larger amount of lean body mass, which mainly consists of skeletal muscle (Marks D. L. et al. Role of the central melanocortin system in cachexia. Cancer Res. (2001) 61: 1432-1438).
  • WO03009847A and WO03009850A describe phenylpiperazinyl-phenylalanine derivatives for the treatment of obesity.
  • Most of the compounds in both patents contain a N-(2-piperidin-4-yl-phenyl)-alkyl, benzyl or aryl sulfonamide group and N-(2-piperazin-4-yl-phenyl)-alkyl, benzyl or aryl sulfonamide group, respectively.
  • These compounds have in common the substituted phenylalanine moiety which is acylated with amino acids, carboxylic acids and sulfonyl chlorides. In some cases the phenylalanine moiety is also alkylated or uraes and urethanes are introduced.
  • Biological data e.g. binding IC 50 or functional activity
  • WO02070511A describes phenylpiperazinyl-phenylalanine amides, phenylpiperidinyl-phenylalanine amides and cyclohexyl-phenylalanine amides as modulators of melanocortin receptors 1 and 4.
  • the phenylalanine amino group is in the most cases acylated with a second amino acid. For amino acids with a basic side chain the amino group can be acylated. Biological data for the compounds are not given.
  • WO02059108A describes melanocortin receptor agonists.
  • the agonists consist of substituted phenylpiperazines which are first acylated with phenylalanines and then with amino acids. Biological data are not provided.
  • WO0074679A describes substituted piperidines as melanocortin-4 receptor agonists.
  • the piperidines are acylated with different substituted phenylalanines, e.g. D-p-chlorophenylalanine, which are subsequently acylated with other amino acids, in particular 1,2,3,4-tetrahydro-isoquinoline-3-carboxylic acid.
  • Example 2 of this patent application binds to the human MC-4-receptor with an IC 50 of 0.92 nM.
  • the compound is acting as an agonist with an EC 50 of 2.1 nM (96% activation).
  • WO9964002A describes spiroindane derivatives as melanocortin receptor agonists.
  • the spiroindanes are acylated with phenylalanine, in particular p-chlorophenylalanine, which is then acylated with unsubstituted and substituted piperazine-2-carboxylic acid, morpholine-3-carboxylic acid and thiomorpholine-3-carboxylic acid, respectively. No biological data is given.
  • WO0170337A describes spiroindane derivatives as melanocortin receptor agonists.
  • the spiroindanes are acylated with phenylalanine, in particular p-chlorophenylalanine, which is then acylated with unsubstituted and substituted 1,2,3,4-tetrahydro-isoquinoline-3-carboxylic acid. No biological data is given.
  • WO0191752A decribes melanocortin receptor agonists.
  • Three examples are described consisting of 3a-benzyl-2-methyl-2,3a,4,5,6,7-hexahydro-pyrazolo[4,3-c]pyridin-3-one which is first acylated with Boc-D-4-chlorophenylalanine following a second acylation step using 1-amino-1,2,3,4-tetrahydro-naphthalene-2-carboxylic acid and 1,2,3,4-tetrahydro-isoquinoline-3-carboxylic acid, respectively. Biological data for the examples is not provided.
  • the present invention relates to novel substituted piperidine and piperazine derivatives of structural formula (I), wherein the variables A, R 1 , Ar, m and n have the meaning as defined below.
  • the present invention also relates to novel substituted piperidine and piperazine derivatives of structural formula (II), wherein the variables A, R 1 , Ar, m and n have a different meaning than in structural formula (I) and are also defined below.
  • the piperidine and piperazine derivatives of structural formulas (I) and (II) are effective as melanocortin receptor modulators and are particularly effective as selective melanocortin-4 receptor (MC-4R) modulators. They are Thereforee useful for the treatment of disorders where the activation or inactivation of the MC-4R are involved.
  • Agonists can be used for the treatment of disorders and diseases such as obesity, diabetes and sexual dysfunction, whereas the antagonists are useful for the treatment of disorders and diseases such as cancer cachexia, muscle wasting, anorexia, anxiety and depression.
  • the present invention also relates to pharmaceutical compositions comprising the compounds of the present invention and a pharmaceutically acceptable carrier.
  • the present invention relates to novel substituted piperidine and piperazine derivatives useful as melanocortin receptor modulators, in particular, selective MC-4R agonists and MC-4R antagonists.
  • the compounds of the present invention are represented by structural formula (I). or a pharmaceutically acceptable salt or a solvate thereof, wherein Ar is:
  • Ar is as defined above, and is preferably aryl, more preferably phenyl or naphthyl. If aryl or heteroaryl are substituted, it is preferably substituted with one to three, more preferably one or two, most preferably one, substituents.
  • the substituents are preferably independently selected from the group consisting of cyano, nitro, perfluoroalkoxy, halo, alkyl, (D)-cycloalkyl, alkoxy and haloalkyl, more preferably perfluoroalkoxy, halo, alkyl, alkoxy or haloalkyl, even more preferably halo, alkyl, alkoxy and/or haloalkyl, in particular halo.
  • Ar is phenyl or naphthyl which both, preferably phenyl, may be substituted with one to three, in particular one, halo, e.g. Cl.
  • the substitution can be in any position, preferably in the 4-position.
  • R 1 is as defined above preferably:
  • A is:
  • R 2 is as defined above. If aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl are substituted, they are preferably independently substituted with one to three, more preferably one substituent selected from the group consisting of oxo, halo, alkyl, N(R 6 ) 2 , OR 6 , SR 6 and/or CO 2 R 6 .
  • R 2 is hydrogen, hydroxy, halo, alkyl, alkoxy, S-alkyl, SO 2 -alkyl, O-alkenyl, S-alkenyl, haloalkyl or (D)-cycloalkyl, more preferably hydrogen, hydroxy, alkoxy, S-alkyl, SO 2 -alkyl, O-alkenyl, S-alkenyl, halo or alkyl, e.g. methyl, ethyl, n-propyl, isopropyl.
  • R 2 is hydrogen, halo, alkyl, haloalkyl, alkoxy, (D)-cycloalkyl, (D)-aryl, (D)-heteroaryl, (D)-heterocyclyl (wherein heterocyclyl excludes a heterocyclyl containing a single nitrogen), and wherein aryl, heteroaryl, heterocyclyl, alkyl and cycloalkyl are substituted or unsubstituted; preferably hydrogen, hydroxy, halo, alkyl, alkoxy, haloalkyl or (D)-cycloalkyl, more preferably hydrogen, alkoxy, halo or alkyl.
  • R 4 and R 5 are each independently as defined above.
  • said ring may contain an additional heteroatom preferably selected from O, S and NR 6 in the ring.
  • alkyl and cycloalkyl are substituted, they are preferably substituted with one to three, more preferably one or two groups independently selected from R 7 and oxo.
  • R 4 and R 5 are each independently preferably selected from the group consisting of hydrogen, alkyl or cycloalkyl; or R 4 and R 5 together with the nitrogen to which they are attached form a 5- to 7-membered ring. More preferably R 4 and R 5 are each independently selected from the group consisting of hydrogen or alkyl, or R 4 and R 5 together with the nitrogen to which they are attached form a 5- to 6-membered ring optionally containing an additional oxygen atom.
  • R 6 is independently hydrogen, alkyl, C(O) alkyl, (D)-aryl or (D)-cycloalkyl, preferably hydrogen, alkyl, C(O) alkyl, (D)-aryl or (D)-cycloalkyl, most preferably hydrogen.
  • R 7 is alkyl, (D)-aryl, (D)-cycloalkyl, (D)-heteroaryl, halo, OR 8 , NHSO 2 R 8 , N(R 8 ) 2 , C ⁇ N, CO 2 R 4 , C(R 8 )(R 8 )N(R 8 ) 2 , nitro, SO 2 N(R 8 ) 2 , S(O) p R 8 , CF 3 or OCF 3 , preferably hydrogen, alkyl, (D)-aryl or (D)-cycloalkyl, more preferably hydrogen.
  • R 8 is as defined above, preferably hydrogen, alkyl or (D)-aryl, more preferably alkyl or (D)-aryl.
  • R 9 is as defined above, preferably hydrogen, alkyl and (D)-cycloalkyl, more preferably alkyl, most preferably methyl, ethyl or tert-butyl.
  • R 10 is as defined above. If aryl and heteroaryl are substituted, they are preferably independently substituted with one to three groups selected from R 7 . Moreover, if alkyl, D, cycloalkyl and heterocyclyl are substituted, they are preferably substituted with one to four groups independently selected from R 7 and oxo. If two R 10 groups together with the atoms to which they are attached form a 5- to 8-membered mono- or bi-cyclic ring system, said ring may contain an additional heteroatom preferably selected from O, S, NR 8 , NBoc and NZ.
  • R 10 is R 9 .
  • R 11 is defined as above, preferably hydrogen, halo, alkyl, alkoxy or C ⁇ N, more preferably hydrogen, halo or C 1 -C 4 -alkyl, most preferably hydrogen.
  • R 12 is as defined above. Moreover, if alkyl, alkoxy, cycloalkyl, aryl, heterocyclyl and heteroaryl are substituted, they are preferably substituted with 1 to 5, more preferably 1 to 3, most preferably 1 or 2 substituents independently selected from R 13 .
  • R 12 is hydrogen, hydroxy, cyano, nitro, halo, alkyl, alkoxy, haloalkyl, (D)-C(O)-heterocyclyl, (D)-C(O)N(R 14 ) 2 , (D)-N(R 14 ) 2 , (D)-NR 14 COR 14 , (D)-NR 14 CON(R 14 ) 2 , (D)-NR 14 C(O)OR 14 , (D)-NR 14 C(R 14 ) ⁇ N(R 14 ), (D)-NR 14 C( ⁇ NR 14 )N(R 14 ) 2 , (D)-NR 14 SO 2 R 14 or (D)-NR 14 SO 2 N(R 14 ) 2 , wherein alkyl or alkoxy are substituted or unsubstituted with one to five, preferably one to three, substituents selected from R 13 .
  • R 12 is cyano, nitro, halo, alkyl, D)-C(O)-heterocyclyl, (D)-N(R 14 ) 2 (D)-NR 14 COR 14 , (D)-NR 14 CON(R 14 ) 2 , (D)-NR 14 C(O)OR 14 or (D)-NR 14 SO 2 R 14 .
  • R 12 is (D)-C(O)-heterocyclyl, (D)-C(O)N(R 14 ) 2 .
  • Halo is preferably F, Cl or Br.
  • R 12 can be on any position of the ring, preferably in the 1-position.
  • R 12 is hydrogen, hydroxy, cyano, nitro, halo, alkyl, alkoxy, haloalkyl, (D)-N(R 14 ) 2 , (D)-NR 14 COR 14 , (D)-NR 14 CON(R 14 ) 2 , (D)-NR 14 C(O)OR 14 , (D)-NR 14 C(R 14 ) ⁇ N(R 14 ), (D)-NR 14 C( ⁇ NR 14 )N(R 14 ) 2 , (D)-NR 14 SO 2 R 14 or (D)-NR 14 SO 2 N(R 14 ) 2 , wherein alkyl or alkoxy are substituted or unsubstituted with one to five, preferably one to three, substituents selected from R 13 .
  • R 12 is cyano, nitro, halo, alkyl, (D)-N(R 14 ) 2 (D)-NR 14 COR 14 , (D)-NR 14 CON(R 14 ) 2 , (D)-NR 14 C(O)OR 14 or (D)-NR 14 SO 2 R 14 .
  • R 13 is independently hydrogen, halo, oxo, N(R 15 ) 2 , alkyl, (D)-cycloalkyl, haloalkyl, alkoxy, heteroaryl, hydroxy or heterocyclyl, wherein heterocyclyl excludes a heterocyclyl containing a single nitrogen, phenyl, (D)-COR 14 , (D)-C(O)OR 14 , (D)-OR 14 , (D)-OCOR 14 , (D)-OCO 2 R 14 , (D)-SR 14 , (D)-SOR 14 or (D)-SO 2 R 14 , wherein aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl is substituted or unsubstituted.
  • aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl are substituted, they are preferably substituted with one to three, preferably one or two, substituents selected from the group consisting of oxo, alkyl, N(R 15 ) 2 , OR 15 , SR 15 and/or CO 2 R 15 .
  • R 13 is hydrogen, halo, alkyl, (D)-cycloalkyl, alkoxy or phenyl, more preferably R 13 is hydrogen, halo, alkyl, alkoxy or phenyl.
  • R 14 is as defined above. If aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl are substituted, they are preferably substituted with one to three, more preferably one or two, substituents selected from the group consisting of oxo, halo, alkyl, N(R 15 ) 2 , OR 15 , SR 15 and/or CO 2 R 15 .
  • R 14 is hydrogen, haloalkyl, alkyl, (D)-cycloalkyl or phenyl, more preferably R 14 is hydrogen, haloalkyl, alkyl or phenyl.
  • R 15 is independently hydrogen, alkyl, C(O)alkyl, aryl or cycloalkyl, preferably hydrogen, alkyl or cycloalkyl, in particular, hydrogen.
  • X is as defined above. If aryl and heteroaryl are substituted, they are preferably substituted with one to three, more preferably one or two, groups selected from R 7 . Moreover, if alkyl, D, cycloalkyl and heterocyclyl are substituted, they are preferably substituted with one to four groups independently selected from R 7 and oxo.
  • X is alkyl, (D)-cycloalkyl, (D)-aryl, (D)-heteroaryl, (D)-heterocyclyl, (D)-NHC(O)R 9 , (D)-CO 2 R 9 or (D)-CON(R 9 R 9 ), more preferably from alkyl, (D)-cycloalkyl, (D)-heterocyclyl, (D)-NHC(O)R 9 or (D)-CON(R 9 R 9 ), most preferably from C 1 -C 4 -alkyl, C 5 -C 7 -cycloalkyl, (D)-CON(R 9 R 9 ) and N-containing heterocyclyl, in particular triazolyl and tetrazolyl.
  • Y is as defined above. If aryl and heteroaryl are substituted, they are preferably substituted with one to three, more preferably one or two, groups selected from R 7 . Moreover if alkyl, D and cycloalkyl are substituted, they are preferably substituted with one to three groups selected from R 7 and oxo.
  • Y is hydrogen, alkyl, (D)-cycloalkyl, (D)-aryl, (D)-heteroaryl or (D)-heterocyclyl, more preferably hydrogen, alkyl, (D)-cycloalkyl or (D)-heterocyclyl, most preferably hydrogen, C 1 -C 4 -alkyl or C 5 -C 7 -cycloalkyl, in particular cyclohexyl.
  • Cy is as defined above, preferably benzene or pyridine, more preferably benzene.
  • D is as defined above, preferably a bond or C 1 -C 4 -alkylene, more preferably a bond or CH 2 .
  • E is as defined above, preferably NSO 2 R 10 , CHN(Y)COR 10 or CHN(Y)SO 2 R 10 , more preferably NSO 2 R 10 .
  • G is as defined above, preferably D or CH-alkyl, more preferably D, in particular CH 2 .
  • J is N or CH
  • T is O or NR 4 , preferably 0; in one embodiment, T is NR 4 ;
  • n 0, 1 or 2, preferably 0 or 1, more preferably 0;
  • n 0, 1 or 2, preferably 0 or 1, more preferably 0;
  • o is 0, 1, 2 or 3, preferably 0, 1 or 2, more preferably 0 or 1;
  • p 0, 1 or 2;
  • q is 0 or 1
  • r is 1 or 2, preferably 1.
  • the variants have the following meanings:
  • Ar is as defined above, and is preferably aryl, more preferably phenyl or naphthyl. If aryl or heteroaryl are substituted, it is preferably substituted with one to three, more preferably one or two, most preferably one, substituents.
  • the substituents are preferably independently selected from the group consisting of: cyano, nitro, perfluoroalkoxy, halo, alkyl, (D)-cycloalkyl, alkoxy and haloalkyl, more preferably cyano, perfluoroalkoxy, halo, alkyl, (D)-cycloalkyl, alkoxy or haloalkyl, even more preferably halo, alkyl, alkoxy and/or haloalkyl, in particular halo.
  • Ar is phenyl or naphthyl which both, preferably phenyl, may be substituted with one to three, in particular one, halo, e.g. Cl.
  • the substitution can be in any position, preferably in the 4-position.
  • R 1 is as defined above, preferably:
  • R 1 is: A is as defined above.
  • R 2 is as defined above. If aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl are substituted, they are preferably substituted with one to three substituents, more preferably one, selected from the group consisting of oxo, alkyl, N(R 3 ) 2 , OR 3 , SR 3 and/or CO 2 R 3 .
  • R 2 is hydrogen, halo, alkyl, haloalkyl, alkoxy or (D)-cycloalkyl, more preferably hydrogen, halo or alkyl, e.g. methyl, ethyl, n-propyl, iso-propyl, most preferably hydrogen.
  • R 3 is as defined above, preferably hydrogen or alkyl, e.g. methyl, ethyl, n-propyl, iso-propyl, more preferably hydrogen.
  • R 4 is as defined above, preferably hydrogen, (D)-aryl, (D)-heteroaryl, (D)-N(R 6 ) 2 , (D)-NR 6 C(O)alkyl, (D)-NR 6 SO 2 alkyl or alkyl, e.g. methyl, ethyl, n-propyl, iso-propyl, more preferably hydrogen.
  • R 5 is as defined above, preferably hydrogen or alkyl, e.g. methyl, ethyl, n-propyl, iso-propyl, more preferably hydrogen.
  • R 6 and R 7 are each independently as defined above.
  • said ring may contain an additional heteroatom preferably selected from O, S and NR 3 in the ring.
  • alkyl and cycloalkyl are substituted, they are preferably substituted with one to three, more preferably one or two, groups independently selected from R 8 and/or oxo.
  • R 6 and R 7 are each independently preferably selected from the group consisting of hydrogen, alkyl or cycloalkyl, or R 6 and R 7 together with the nitrogen to which they are attached form a 5- to 7-membered ring. More preferably R 6 and R 7 are each independently selected from the group consisting of hydrogen or alkyl, or R 6 and R 7 together with the nitrogen to which they are attached form a 5- to 6-membered ring optionally containing an additional oxygen atom.
  • R 8 is alkyl, (D)-aryl, (D)-cycloalkyl, (D)-heteroaryl, halo, OR 9 , NHSO 2 R 9 , N(R 9 ) 2 , C ⁇ N, CO 2 R 6 , C(R 9 )(R 9 )N(R 9 ) 2 , nitro, SO 2 N(R 9 ) 2 , S(O) u R 9 , CF 3 or OCF 3 .
  • R 8 is alkyl, OR 9 , (D)-aryl, (D)-cycloalkyl, (D)-heteroaryl or halo.
  • R 9 is as defined above, preferably hydrogen, (D)-aryl or alkyl, e.g. methyl, ethyl, n-propyl, iso-propyl, more preferably hydrogen or (D)-aryl.
  • R 10 is as defined above, preferably hydrogen or C 1 -C 4 alkyl as defined below, more preferaby hydrogen, methyl or ethyl, most preferably hydrogen or methyl.
  • R 11 is as defined above, preferably hydrogen or alkyl, more preferably hydrogen or C 1 -C 6 alkyl as defined below, in particular C 1 -C 5 alkyl, e.g. methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl and n-pentyl, most preferably hydrogen or tert-butyl.
  • R 12 is as defined above. If aryl and heteroaryl are substituted, they are preferably substituted with one to three, more preferably one or two, groups selected from R 7 . Moreover, if alkyl, D, cycloalkyl and heterocyclyl are substituted, they are preferably substituted with one to four, more preferably one or two, groups independently selected from R 8 and/or oxo. Furthermore, if two R 12 groups together with the atoms to which they are attached form a 5- to 8-membered mono- or bi-cyclic ring system, said ring may contain an additional heteroatom preferably selected from O, S, NR 9 , NBoc and NZ.
  • R 12 is R 11 .
  • R 13 is as defined above, preferably hydrogen, halo, alkyl, alkoxy and C ⁇ N, more preferably hydrogen, methyl or ethyl, most preferably hydrogen or methyl.
  • R 14 is as defined above. Moreover, if alkyl, alkoxy, cycloalkyl, aryl, heterocyclyl and heteroaryl are substituted, they are preferably substituted with 1 to 5, more preferably 1 to 3, most preferably 1 or 2, substituents independently selected from R 15 .
  • R 14 is hydrogen, hydroxy, cyano, nitro, halo, alkyl, alkoxy, haloalkyl, (D)-C(O)-heterocyclyl, (D)-C(O)N(R 16 ) 2 , (D)-N(R 16 ) 2 , (D)-NR 16 COR 14 , (D)-NR 14 CON(R 14 ) 2 , (D)-NR 14 C(O)OR 16 , (D)-NR 16 C(R 16 ) ⁇ N(R 16 ), (D)-NR 16 C( ⁇ NR 16 )N(R 16 ) 2 , (D)-NR 16 SO 2 R 16 or (D)-NR 16 SO 2 N(R 16 ) 2 , wherein alkyl or alkoxy are substituted or unsubstituted with one to five, preferably one to three, substituents selected from R 15 .
  • R 14 is cyano, nitro, halo, alkyl, (D)-C(O)-heterocyclyl, (D)-C(O)N(R 16 ) 2 , (D)-N(R 16 ) 2 , (D)-NR 16 COR 16 , (D)-NR 16 CON(R 16 ) 2 , (D)-NR 16 C(O)OR 16 or (D)-NR 16 SO 2 R 16 .
  • R 16 is (D)-C(O)-heterocyclyl, (D)-C(O)N(R 16 ) 2 or NR 16 COR 16 .
  • Halo is preferably F, Cl or Br.
  • R 14 can be on any position of the ring, preferably in the 1-position.
  • R 14 is hydrogen, hydroxy, cyano, nitro, halo, alkyl, alkoxy, haloalkyl, (D)-N(R 16 ) 2 , (D)-NR 16 COR 14 , (D)-NR 14 CON(R 14 ) 2 , (D)-NR 14 C(O)OR 16 , (D)-NR 16 C(R 16 ) ⁇ N(R 16 ), (D)-NR 16 C( ⁇ NR 16 )N(R 16 ) 2 , (D)-NR 16 SO 2 R 16 or (D)-NR 16 SO 2 N(R 16 ) 2 , wherein alkyl or alkoxy are substituted or unsubstituted with one to five, preferably one to three, substituents selected from R 15 .
  • R 14 is cyano, nitro, halo, alkyl, (D)-N(R 16 ) 2 , (D)-NR 16 COR 16 , (D)-NR 16 CON(R 16 ) 2 , (D)-NR 16 C(O)OR 16 or (D)-NR 16 SO 2 R 16 .
  • R 15 is independently hydrogen, halo, oxo, N(R 3 ) 2 , alkyl, (D)-cycloalkyl, alkoxy, haloalkyl, heteroaryl, hydroxy, heterocyclyl, wherein heterocyclyl excludes a heterocyclyl containing a single nitrogen, phenyl, (D)-COR 16 , (D)-C(O)OR 16 , (D)-R 16 , (D)-COR 16 , (D)-OCO 2 R 16 , (D)-SR 16 , (D)-SOR 16 or (D)-SO 2 R 16 , wherein aryl, heteroaryl, heterocyclyl, alkyl and cycloalkyl are substituted or unsubstituted.
  • aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl are substituted, they are preferably substituted with one to three, more preferably one or two, substituents selected from the group consisting of oxo, alkyl, N(R 16 ) 2 , OR 16 , SR 16 and/or CO 2 R 16 .
  • R 15 is hydrogen, halo, alkyl, (D)-cycloalkyl, alkoxy or phenyl, more preferably R 15 is hydrogen, halo, alkyl, alkoxy or phenyl.
  • R 16 is as defined above. If aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl are substituted, they are preferably substituted with one to three, more preferably one or two, substituents selected from the group consisting of oxo, alkyl, N(R 3 ) 2 , OR 3 , SR 3 and/or CO 2 R 3 .
  • R 16 is hydrogen, halo, alkyl, (D)-cycloalkyl, alkoxy or phenyl, more preferably R 16 is hydrogen, halo, alkyl, alkoxy or phenyl.
  • Cy is as defined above wherein heteroaryl and heterocyclyl are preferably 5- or 6-membered and carbocyclyl is preferably 5- or 7-membered.
  • Cy is aryl, more preferably benzene.
  • X is as defined above. If aryl and heteroaryl are substituted, they are preferably substituted with one to three, more preferably one or two, groups selected from R 8 . Moreover, if alkyl, D, cycloalkyl and heterocyclyl are substituted, they are preferably substituted with one to four groups independently selected from R 8 and/or oxo.
  • X is alkyl, (D)-cycloalkyl, (D)-aryl, (D)-heteroaryl, (D)-heterocyclyl, (D)-NHC(O)R 11 , (D)-CO 2 R 11 or (D)-CON(R 11 R 11 ), more preferably alkyl, (D)-cycloalkyl, (D)-heterocyclyl, (D)-NHC(O)R 1 , or (D)-CON(R 11 R 11 ), most preferably C 1 -C 4 -alkyl, C 5 -C 7 -cycloalkyl, (D)-CON(R 11 R 11 ) and N-containing heterocyclyl, in particular triazolyl and tetrazolyl.
  • Y is as defined above. If aryl and heteroaryl are substituted, they are preferably substituted with one to three, more preferably one or two, groups selected from R 8 . Moreover, if alkyl, D and cycloalkyl are substituted, they are preferably substituted with one to three groups selected from R 8 and/or oxo.
  • Y is hydrogen, alkyl, (D)-cycloalkyl, (D)-aryl, (D)-heteroaryl or (D)-heterocyclyl, more preferably alkyl, (D)-cycloalkyl or (D)-heterocyclyl, most preferably hydrogen, C 1 -C 4 -alkyl and C 5 -C 7 -cycloalkyl, in particular cyclohexyl.
  • Cy′ is as defined above, preferably benzene or pyridine, more preferably benzene.
  • D is as defined above, preferably a bond or C 1 -C 4 alkylene, more preferably a bond or CH 2 .
  • E is as defined above, preferably NSO 2 R 10 , CHN(Y)COR 12 or CHN(Y)SO 2 R 12 , more preferably NSO 2 R 10 .
  • G is as defined above, preferably D or CH alkyl, more preferably D, in particular CH 2 .
  • L is as defined above, preferably NR 5 ;
  • M is as defined above, preferably bond or CH 2 ;
  • n 0, 1 or 2, preferably 0 or 1, unless m is 0, then n is 1;
  • o 0, 1, 2 or 3, preferably 0, 1 or 2, more preferably 1;
  • p is 0 or 1, preferably 0;
  • q is 0 or 1, preferably 0;
  • r is 1 or 2, preferably 1;
  • s is 0, 1, 2, 3, 4 or 5, preferably 0, 1, 2 or 3, more preferably 0 or 1, most preferably 0;
  • u 0, 1 or 2;
  • v 0 or 1.
  • Aryl is an aromatic mono- or polycyclic moiety with 6 to 20 carbon atoms which is preferably selected from phenyl, biphenyl, naphthyl, tetrahydronaphthyl, fluorenyl, indenyl or phenanthrenyl, more preferably from phenyl or naphthyl.
  • Heteroaryl is an aromatic moiety having 6 to 20 carbon atoms with at least one heterocycle and is preferably selected from thienyl, benzothienyl, naphthothienyl, furanyl, benzofuranyl, chromenyl, indolyl, isoindolyl, indazolyl, quinolyl, isoquinolyl, phthalazinyl, quinoxalinyl, cinnolinyl or quinazolinyl, more preferably from thienyl, furanyl, benzothienyl, benzofuranyl or indolyl.
  • Heterocyclyl is a saturated, unsaturated or aromatic ring containing at least one heteroatom selected from O, N and/or S and 1 to 6 carbon atoms and is preferably selected from azetidin-2-one-1-yl, pyrrolidin-2-one-1-yl, piperid-2-one-1-yl and azepan-2-one-1-yl, thienyl, furyl, piperidinyl, pyranyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, isothiazolyl or isoxazyl, more preferably from pyridyl, piperidinyl, imidazolyl or pyrazinyl.
  • Carbocyclyl is a monocyclic or polycyclic ring system of 3 to 20 carbon atoms which may be saturated, unsaturated or aromatic.
  • Alkyl is straight chain or branched alkyl having preferably 1 to 8 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, neopentyl, hexyl or heptyl, more preferably 1 to 4 carbon atoms.
  • Cycloalkyl is an alkyl ring having preferably 3 to 8 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl, more preferably 3 to 6 carbon atoms.
  • Alkenyl is straight chain or branched alkenyl having preferably 2 to 8 carbon atoms such as vinyl, allyl, methallyl, buten-2-yl, buten-3-yl, penten-2-yl, penten-3-yl, penten-4-yl, 3-methyl-but-3-enyl, 2-methyl-but-3-enyl, 1-methyl-but-3-enyl, hexenyl or heptenyl, more preferably 2 to 4 atoms.
  • Alkoxy is O-alkyl wherein alkyl is as defined above and has preferably 1 to 4 carbon atoms, preferably 1 or 3 carbon atoms.
  • Halo or halogen is a halogen atom preferably selected from F, Cl, Br and I, preferably F, Cl and Br.
  • Haloalkyl is an alkyl moiety as defined above having preferably 1 to 4 carbon atoms, more preferably 1 or 2 carbon atoms, wherein at least one, preferably 1 to 3 hydrogen atoms, have been replaced by a halogen atom.
  • Preferred examples are —CF 3 , —CH 2 CF 3 and —CF 2 CF 3 .
  • the alkylene moiety may be a straight chain or branched chain group.
  • Said alkylene moiety preferably has 1 to 6 carbon atoms. Examples thereof include methylene, ethylene, n-propylene, n-butylene, n-pentylene, n-hexylene, iso-propylene, sec.-butylene, tert.-butylene, 1,1-dimethyl propylene, 1,2-dimethyl propylene, 2,2-dimethyl propylene, 1,1-dimethyl butylene, 1,2-dimethyl butylene, 1,3-dimethyl butylene, 2,2-dimethyl butylene, 2,3-dimethyl butylene, 3,3-dimethyl butylene, 1-ethyl butylene, 2-ethyl butylene, 3-ethyl butylene, 1-n-propyl propylene, 2-n-propyl propylene, 1-iso-propyl propylene, 2-iso-propyl prop
  • the compounds of structural formulas (I) and (II) are effective as melanocortin receptor modulators and are particularly effective as selective modulators of MC-4R. They are thereforee useful for the treatment and/or prevention of disorders responsive to the activation and inactivation of MC-4R, such as cancer cachexia, muscle wasting, anorexia, anxiety, depression, obesity, diabetes, sexual dysfunction and other diseases with MC-4R involvement.
  • Some of the compounds described herein may exist as tautomers, such as keto-enol tautomers.
  • the individual tautomers, as well as mixtures thereof, are encompassed within the compounds of structural formulas (I) and (II).
  • Compounds of structural formulas (I) and (II) may be separated into their individual diastereoisomers by, for example, fractional crystallization from a suitable solvent, for example methanol or ethyl acetate or a mixture thereof, or via chiral chromatography using an optically active stationary phase.
  • Absolute stereochemistry may be determined by X-ray crystallography of crystalline products or crystalline intermediates which are derivatized, if necessary, with a reagent containing an asymmetric center of known absolute configuration.
  • any stereoisomer of a compound of the general formulas (I) and (II) may be obtained by stereospecific synthesis using optically pure starting materials or reagents of known absolute configuration.
  • salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc and the like. Particularly preferred are the ammonium, calcium, lithium, magnesium, potassium and sodium salts.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N′-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like.
  • basic ion exchange resins such as arginine
  • salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids.
  • acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, formic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, malonic, mucic, nitric, pamoic, pantothenic, phosphoric, propionic, succinic, sulfuric, tartaric, ptoluenesulfonic, trifluoroacetic acid and the like.
  • Particularly preferred are citric, fumaric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric and tartaric acids.
  • Compounds of formulas (I) and (II) are melanocortin receptor modulators and as such are useful in the treatment, control or prevention of diseases, disorders or conditions responsive to the activation or inactivation of one or more of the melanocortin receptors including, but not limited to, MC-1R, MC-2R, MC-3R, MC-4R or MC-5R.
  • Such diseases, disorders or conditions include, but are not limited to, cancer cachexia, muscle wasting, anorexia, anxiety, depression, obesity (by reducing appetite, increasing metabolic rate, reducing fat intake or reducing carbohydrate craving), diabetes mellitus (by enhancing glucose tolerance, decreasing insulin resistance), hypertension, hyperlipidemia, osteoarthritis, cancer, gall bladder disease, sleep apnea, depression, anxiety, compulsion, neuroses, insomnia/sleep disorder, substance abuse, pain, male and female sexual dysfunction (including impotence, loss of libido and erectile dysfunction), fever, inflammation, immune-modulation, rheumatoid arthritis, skin tanning, acne and other skin disorders, neuroprotective and cognitive and memory enhancement including the treatment of Alzheimer's disease.
  • Some compounds encompassed by formulas (I) and (II) show highly selective affinity for the melanocortin-4 receptor relative to MC-1R, MC-2R, MC-3R and MC-5R, which makes them especially useful in the prevention and treatment of cancer cachexia, muscle wasting, anorexia, anxiety, depression, obesity, as well as male and/or female sexual dysfunction, including erectile dysfunction.
  • “Male sexual dysfunction” includes impotence, loss of libido and erectile dysfunction.
  • “Female sexual dysfunction” can be seen as resulting from multiple components including dysfunction in desire, sexual arousal, sexual receptivity and orgasm.
  • Any suitable route of administration may be employed for providing a mammal, especially a human with an effective dosage of a compound of the present invention.
  • oral, rectal, topical, parenteral, ocular, pulmonary, nasal and the like may be employed.
  • Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, aerosols and the like.
  • compounds of formulas (I) and (II) are administered orally or topically.
  • the effective dosage of active ingredient employed may vary depending on the particular compound employed, the mode of administration, the condition being treated and the severity of the condition being treated. Such dosage may be ascertained readily by a person skilled in the art.
  • the compounds of the present invention are administered at a daily dosage of from about 0.001 milligram to about 100 milligrams per kilogram of body weight, preferably given in a single dose or in divided doses two to six times a day, or in sustained release form.
  • the total daily dose will generally be from about 0.07 milligrams to about 3500 milligrams. This dosage regimen may be adjusted to provide the optimal therapeutic response.
  • the compounds of the present invention are administered at a daily dosage of from about 0.001 milligram to about 100 milligrams per kilogram of body weight, preferably given in a single dose or in divided doses two to six times a day, or in sustained release form.
  • the total daily dose will generally be from about 0.07 milligrams to about 3500 milligrams. This dosage regimen may be adjusted to provide the optimal therapeutic response.
  • the compounds of the present invention are administered at a daily dosage of from about 0.001 milligram to about 100 milligram per kilogram of animal body weight, preferably given in a single dose or in divided doses two to six times a day, or in sustained release form.
  • the total daily dose will generally be from about 0.07 milligrams to about 3500 milligrams. This dosage regimen may be adjusted to provide the optimal therapeutic response.
  • compounds of the present invention are given in a dose range of 0.001 milligram to about 100 milligram per kilogram of body weight, preferably as a single dose orally or as a nasal spray.
  • the compound of formulas (I) and (II) is preferably formulated into a dosage form prior to administration. Accordingly the present invention also includes a pharmaceutical composition comprising a compound of formulas (I) and (II) and a suitable pharmaceutical carrier.
  • the active ingredient (a compound of formulas (I) and (II)) is usually mixed with a carrier, or diluted by a carrier, or enclosed within a carrier, which may be in the form of a capsule, sachet, paper or other container.
  • a carrier which may be in the form of a capsule, sachet, paper or other container.
  • the carrier serves as a diluent, it may be a solid, semisolid or liquid material which acts as a vehicle, excipient or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosol (as a solid or in a liquid medium), soft and hard gelatin capsules, suppositories, sterile injectable solutions and sterile packaged powders.
  • Suitable carriers, excipients and diluents include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water syrup, methyl cellulose, methyl and propylhydroxybenzoates, talc, magnesium stearate and mineral oil.
  • the formulations can additionally include lubricating agents, wetting agents, emulsifying and suspending agents, preserving agents, sweetening agents or flavoring agents.
  • the compositions of the invention may be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient.
  • Preparation of the compounds of the present invention may be carried out via sequential or convergent synthetic routes.
  • the skilled artisan will recognize that, in general, the three moieties of a compound of formulas (I) and (II) are connected via amide bonds.
  • the skilled artist can, Thereforee, readily envision numerous routes and methods of connecting the three moieties via standard peptide coupling reaction conditions.
  • standard peptide coupling reaction conditions means coupling a carboxylic acid with an amine using an acid activating agent such as EDC, dicyclohexylcarbodiimide and benzotriazol-1-yloxytris(dimethylamino)-phosphonium hexafluorophosphate, in a inert solvent such as DCM, in the presence of a catalyst such as HOBt.
  • an acid activating agent such as EDC, dicyclohexylcarbodiimide and benzotriazol-1-yloxytris(dimethylamino)-phosphonium hexafluorophosphate
  • a catalyst such as HOBt.
  • Protecting groups like Z, Boc and Fmoc are used extensively in the synthesis, and their removal conditions are well known to those skilled in the art.
  • removal of Z groups can he achieved by catalytic hydrogenation with hydrogen in the presence of a noble metal or its oxide, such as palladium on activated carbon in a protic solvent, such as ethanol.
  • a protic solvent such as ethanol.
  • removal of Z can also be achieved by treatment with a solution of hydrogen bromide in acetic acid, or by treatment with a mixture of TFA and dimethylsulfide.
  • Removal of Boc protecting groups is carried out in a solvent such as methylene chloride, methanol or ethyl acetate with a strong acid, such as TFA or HCl or hydrogen chloride gas.
  • the compounds of formulas (I) and (II), when existing as a diastereomeric mixture, may be separated into diastereomeric pairs of enantiomers by fractional crystallization from a suitable solvent such as methanol, ethyl acetate or a mixture thereof.
  • the pair of enantiomers thus obtained may be separated into individual stereoisomers by conventional means by using an optically active acid as a resolving agent.
  • any enantiomer of a compound of the formulas (I) and (II) may be obtained by stereospecific synthesis using optically pure starting materials or reagents of known configuration.
  • the compounds of formulas (I) and (II) of the present invention can be prepared according to the procedures of the following Schemes and Examples, using appropriate materials and are further exemplified by the following specific examples. Moreover, by utilizing the procedures described herein, in conjunction with ordinary skills in the art, additional compounds of the present invention claimed herein can be readily prepared.
  • the compounds illustrated in the examples are not, however, to be construed as forming the only genus that is considered as the invention.
  • the Examples further illustrate details for the preparation of the compounds of the present invention. Those skilled in the art will readily understand that known variations of the conditions and processes of the following preparative procedures can be used to prepare these compounds.
  • the instant compounds are generally isolated in the form of their pharmaceutically acceptable salts, such as those described previously.
  • the free amine bases corresponding to the isolated salts can be generated by neutralization with a suitable base, such as aqueous sodium hydrogencarbonate, sodium carbonate, sodium hydroxide and potassium hydroxide, and extraction of the liberated amine free base into an organic solvent followed by evaporation.
  • a suitable base such as aqueous sodium hydrogencarbonate, sodium carbonate, sodium hydroxide and potassium hydroxide
  • the amine free base isolated in this manner can be further converted into another pharmaceutically acceptable salt by dissolution in an organic solvent followed by addition of the appropriate acid and subsequent evaporation, precipitation or crystallization. All temperatures are degrees Celsius.
  • Mass spectra (MS) were measured by electron-spray ion-mass spectroscopy.
  • an appropriate “A moiety” e.g., 4-cyclohexyl-4-[1,2,4]triazol-1-yl-methyl-piperidine
  • B moiety e.g., L-Boc-p-Cl-Phe-OH
  • the coupled AB compound is then coupled to an appropriate “C moiety” followed by deprotection of Boc group and salt formation.
  • C moiety is not protected with Boc group, the final compound can be obtained without the deprotection step.
  • an appropriate “AB moiety” is coupled to an appropriate “C moiety” in the presence of EDC/HOBt followed by deprotection of Boc group and salt formation.
  • C moiety is not protected with Boc group, the final compound can be obtained without the deprotection step.
  • an appropriate “BC moiety” is coupled to an appropriate “A moiety” in the presence of EDC/HOBt followed by deprotection of Boc group and salt formation.
  • C moiety is not protected with Boc group, the final compound can be obtained without the deprotection step.
  • EDC/HOAt for coupling of A with Boc-B—OH, EDC/HOAt, EDC/HOBt or DCC/HOBt can be used.
  • the starting material of Boc-protected piperazine or piperidine (A moiety) can be deprotected in the presence of TFA/CH 2 Cl 2 , HCl/EtOAc, HCl/dioxane or HCl in MeOH/Et 2 O with or without a cation scavenger, such as dimethyl sulfide (DMS) before being subjected to the coupling procedure. It can be free-based before being subjected to the coupling procedure or in some cases used as the salt.
  • a cation scavenger such as dimethyl sulfide (DMS)
  • a suitable solvent such as CH 2 Cl 2 ; DMF, THF or a mixture of the above solvents, can be used for the coupling procedure.
  • a suitable base includes triethylamine (TEA), diisopropyethylamine (DIPEA), N-methymorpholine (NMM), collidine or 2,6-lutidine.
  • a base may not be needed when EDC/HOBt is used.
  • the reaction mixture can be diluted with an appropriate organic solvent, such as EtOAc, CH 2 Cl 2 or Et 2 O, which is then washed with aqueous solutions, such as water, HCl, NaHSO 4 , bicarbonate, NaH 2 PO 4 , phosphate buffer (pH 7), brine or any combination thereof.
  • an appropriate organic solvent such as EtOAc, CH 2 Cl 2 or Et 2 O
  • aqueous solutions such as water, HCl, NaHSO 4 , bicarbonate, NaH 2 PO 4 , phosphate buffer (pH 7), brine or any combination thereof.
  • the reaction mixture can be concentrated and then be partitioned between an appropriate organic solvent and an aqueous solution.
  • the reaction mixture can be concentrated and subjected to chromatography without aqueous workup.
  • Protecting groups such as Boc, Z, Fmoc and CF 3 CO can be deprotected in the presence of H 2 /Pd—C, TFA/DCM, HCl/EtOAc, HCl/dioxane, HCl in MeOH/Et 2 O, NH 3 /MeOH or TBAF, with or without a cation scavenger, such as thioanisole, ethane thiol and dimethyl sulfide (DMS).
  • the deprotected amines can be used as the resulting salt or are free-based by dissolving in DCM and washing with aqueous bicarbonate or aqueous NaOH.
  • the deprotected amines can also be free-based by ion exchange chromatography.
  • the “A moieties” can be prepared as described in the corresponding literature: 4-Cyclohexyl-4-[1,2,4]triazol-1-ylmethyl-piperidine (WO0074679), 4-cyclohexyl-piperidine-4-carboxylic acid tert-butylamide (WO0170708), 1,2-dihydro-1-methanesulfonylspiro-[3H-indole-3,4′-piperidine] (US005536716), 3a-benzyl-2-methyl-2,3a,4,5,6,7-hexahydro-pyrazolo[4,3-c]pyridin-3-one (WO9858949) and N-(2-piperazin-1-yl-benzyl)-methane-sulfonamide, 1-(2-[1,2,4]triazol-1-ylmethyl-phenyl)-piperazine, N-(2-piperazin-1-yl-phenyl)-isobutyr
  • N-(2-piperazin-1-yl-phenyl)-benzamide (4-methyl-piperazin-1-yl)-(2-piperazin-1-yl-phenyl)-methanone, morpholin-4-yl-(2-piperazin-1-yl-phenyl)-methanone, (2-piperazin-1-yl-phenyl)-pyrrolidin-1-yl-methanone, N-(2-methoxy-ethyl)-2-piperazin-1-yl-benzamide, N-(2-dimethylamino-ethyl)-2-piperazin-1-yl-benzamide, 2-piperazin-1-yl-N-(2,2,2-trifluoro-ethyl)-benzamide, N-cyclopropyl-2-piperazin-1-yl-benzamide, 4-methyl-N-(2-piperazin-1-yl-phenyl)-benzene-sulfonamide, 4-bromo-2-piperazin-1-yl-
  • ethyl 3-bromo-4-oxochromene-2-carboxylate 1 J. Chem. Soc. Perkin Trans. I 1986, 1643-1649
  • amines can be reacted with amines, with or without a base such as K 2 CO 3 , in an appropriate solvent such as MeCN, to form products 2 which are subsequently treated with a reagent such as HBr/HOAc, to form carboxylic acids 3.
  • R 8 is hydrogen
  • the free amine can be protected with a reagent such as Boc 2 O in the presence of TEA and DMAP in an appropriate solvent.
  • substituted phenols 7 can be reacted with triethylamine followed by dimethyl acetylendicarboxylate in diethyl ether to yield compounds 8 ( Aust. J. Chem. 1995, 48, 677-686). Saponification of the latter with aqueous sodium hydroxide leads to acids 9 which are subsequently cyclized to the chromone-2-carboxylic acids 10 using concentrated sulfuric acid in acetyl chloride.
  • methoxy-substituted chromone-2-carboxylic acids can be demethylates with reagents such as hydroiodic acid in an appropriate solvent such as glacial acetic acid to yield the corresponding hydroxy-substituted chromone-2-carboxylic acids.
  • 6-ethylchromone-2-carboxylic acid 6-isopropylchromone-2-carboxylic acid, 6-methoxychromone-2-carboxylic acid, 6-trifluoromethylchromone-2-carboxylic acid, 6-tert.-butylchromone-2-carboxylic acid, 6-chlorochromone-2-carboxylic acid, 6-trifluoromethoxychromone-2-carboxylic acid, 8-methoxychromone-2-carboxylic acid, 6-trifluoromethylsulfanylchromone-2-carboxylic acid, 8-chlorochromone-2-carboxylic acid, 8-fluorochromone-2-carboxylic acid 7-chlorochromone-2-carboxylic acid, 6-ethoxychromone-2-carboxylic acid, 6-methanesulfonylchromone-2-carboxylic acid, 8-oxo-8H-[1,3]dioxolo[4,5
  • the intermediate was dissolved in ethanol (100 ml) and heated at 100° C. for 15 min; concentrated HCl (2 ml) was added, and the solution stirred at 100° C. for 1.5 h. Immediately after addition of the acid a precipitate was formed. After cooling to room temperature the reaction mixture was diluted with water (150 ml) and the pale yellow precipitate was filtered off and washed with water. The product was dried under reduced pressure.
  • chromone-2-carboxylic acids were prepared using method 2: 6-methoxychromone-2-carboxylic acid, 7-methoxychromone-2-carboxylic acid, 6,7-dimethylchromone-2-carboxylic acid, 6,7-dimethoxychromone-2-carboxylic acid, 6-chlorochromone-2-carboxylic acid, 6,8-difluorochromone-2-carboxylic acid, 6,8-dichlorochromone-2-carboxylic acid and 7-fluorochromone-2-carboxylic acid.
  • chromone-2-carboxylic acids were prepared using the demethylation method: 6-hydroxychromone-2-carboxylic acid, 7-hydroxychromone-2-carboxylic acid, 8-hydroxychromone-2-carboxylic acid, 6,7-dihydroxychromone-2-carboxylic acid and 6-hydroxy-7-methoxychromone-2-carboxylic acid.
  • a membrane binding assay is used to identify competitive inhibitors of fluorescence labeled NDP-alpha-MSH binding to HEK293 cell membrane preparations expressing human melanocortin receptors.
  • test compound or unlabeled NDP-alpha-MSH is dispensed at varying concentrations to a 384 well microtiter plate. Fluorescence labeled NDP-alpha-MSH is dispensed at a single concentration, followed by addition of membrane preparations. The plate is incubated for 5 h at room temperature.
  • the degree of fluorescence polarization is determined with a fluorescence polarization microplate reader.
  • a functional cellular assay based on competition between unlabeled CAMP and a fixed quantity of fluorescence labeled cAMP for a limited number of binding sites on a cAMP specific antibody, is used to discriminate melanocortin receptor agonists from antagonists by fluorescence polarization.
  • HEK293 cells expressing one of the human melanocortin receptors are transferred to 384 well microtiter plates, an appropriate amount of cAMP antibody is added, followed by the addition of different concentrations of the test compound to effect cAMP production.
  • Cells are lysed and a fluorescence labeled cAMP conjugate is dispensed.
  • the plate is read on a fluorescence polarization microplate reader and the amount of cAMP produced as a response to a test compound is compared to the production of cAMP resulting from stimulation with NDP-alpha-MSH.
  • the compound is dispensed at different concentrations to cells stimulated by an appropriate amount of NDP- ⁇ -MSH. Inhibition of cAMP production is determined by comparing the inhibition of cAMP production of the test compound to the inhibition of cAMP production by a known inhibitor tested at the same concentrations.
  • Food intake in rats is measured after i.p. or p.o. administration of the test compound (see e.g. Chen, A. S. et al. Transgenic Res 2000 April; 9(2):145-54).
  • LPS lipopolysaccharide
  • This conditioning takes about 4 days. Day 1, the animals are placed in a darkened restrainer and left for 15-30 minutes. Day 2, the animals are restrained in a supine position in the restrainer for 15-30 minutes. Day 3, the animals are restrained in the supine position with the penile sheath retracted for 15-30 minutes. Day 4, the animals are restrained in the supine position with the penile sheath retracted until penile responses are observed. Some animals require additional days of conditioning before they are completely acclimated to the procedures; non-responders are removed from further evaluation. After any handling or evaluation, animals are given a treat to ensure positive reinforcement.
  • Rats are gently restrained in a supine position with their anterior torso placed inside a cylinder of adequate size to allow for normal head and paw grooming.
  • the diameter of the cylinder is approximately 8 cm.
  • the lower torso and hind limbs are restrained with a nonadhesive material (vetrap).
  • An additional piece of vetrap with a hole in it, through which the glans penis will be passed, is fastened over the animal to maintain the preputial sheath in a retracted position.
  • Penile responses will be observed, typically termed ex copula genital reflex tests. Typically, a series of penile erections will occur spontaneously within a few minutes after sheath retraction.
  • the types of normal reflexogenic erectile responses include elongation, engorgement, cup and flip.
  • An elongation is classified as an extension of the penile body.
  • Engorgement is a dilation of the glans penis.
  • a cup is defined as an intense erection where the distal margin of the glans penis momentarily flares open to form a cup.
  • a flip is a dorsiflexion of the penile body.
  • Baseline and or vehicle evaluations are conducted to determine how, and if, an animal will respond. Some animals have a long duration until the first response while others are non-responders altogether. During this baseline evaluation latency to first response, number and type of responses are recorded. The testing time frame is 15 minutes after the first response.
  • test compound After a minimum of 1 day between evaluations, these same animals are administered the test compound at 20 mg/kg and evaluated for penile reflexes. All evaluations are videotaped and scored later. Data are collected and analyzed using paired 2 tailed t-tests to compared baseline and/or vehicle evaluations to drug treated evaluations for individual animals. Groups of a minimum of 4 animals are utilized to reduce variability.
  • mice can be dosed by a number of routes of administration depending on the nature of the study to be performed.
  • the routes of administration includes intravenous (IV), intraperitoneal (IP), subcutaneous (SC) and intracerebral ventricular (ICV).
  • Rodent assays relevant to female sexual receptivity include the behavioral model of lordosis and direct observations of copulatory activity. There is also a urethrogenital reflex model in anesthetized spinally transected rats for measuring orgasm in both male and female rats. These and other established animal models of female sexual dysfunction are described in McKenna K E et al, A Model For The Study of Sexual Function In Anesthetized Male And Female Rats, Am. J. Physiol. (Regulatory Integrative Comp. Physiol 30): R1276-R1285, 1991; McKenna K E et al, Modulation By Peripheral Serotonin of The Threshold For sexual Reflexes In Female Rats, Pharm. Bioch.
  • representative compounds of the present invention bind to the human melanocortin-4 receptor.
  • Representative compounds of the present invention were also tested in the functional assay and found to be non-activating or activating the melanocortin-4 receptor with high EC 50 values and low stimulation.
  • the corresponding compound with D-Tic as “C moiety” (WO0074679, example 2) is described to bind to the melanocortin-4 receptor with an IC 50 between 0.92 and 7 nM and to act as a full agonist of the melanocortin-4 receptor with an EC 50 between 2.1 and 4 nM (WO0074679, example 2 ; J. Med.
  • representative compounds of the present invention are also active as antagonists when tested in vivo.
  • Example 15 is active in the spontaneous feeding paradigm at a dose of 3 mg/kg when administered orally.
  • the test animals show a significantly increase in food intake.
  • the corresponding compound with D-Tic as “C moiety” is reported to act as in vivo agonist in a conscious rat model of reflexogenic erections when administered i.v. or p.o. (WO0074679, example 2) and it was also evaluated for its effects oh food intake ( J. Med. Chem. 2002, 45, 4589-4593, compound 1). Food intake was significantly reduced on administration of the compound.
  • Example 66 is active in the spontaneous feeding paradigm as well as in the model of LPS-induced cachexia.
  • the test animals show a significant increase in food intake at dose of 3 mg/kg p.o. and in the acute model of lipopolysaccharide-induced cachexia the test animals show a significant increase in cumulative food intake at a dose of 10 mg/kg p.o.
  • Example 66 As a specific embodiment of an oral composition of a compound of the present invention, 25 mg of Example 66 is formulated with sufficient finely divided lactose to provide a total amount of 580 to 590 mg to fill a size 0 hard gelatin capsule.
  • Example 113 is formulated with sufficient finely divided lactose to provide a total amount of 580 to 590 mg to fill a size 0 hard gelatin capsule.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Diabetes (AREA)
  • Neurology (AREA)
  • Endocrinology (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Pain & Pain Management (AREA)
  • Neurosurgery (AREA)
  • Reproductive Health (AREA)
  • Nutrition Science (AREA)
  • Gynecology & Obstetrics (AREA)
  • Emergency Medicine (AREA)
  • Psychiatry (AREA)
  • Child & Adolescent Psychology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Hydrogenated Pyridines (AREA)
  • Pyrane Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)

Abstract

The present invention relates to novel substituted piperidine and piperazine derivatives as melanocortin-4 receptor (MC-4R) modulators. MC-4R agonists of the invention can be used for the treatment of disorders and diseases such as obesity, diabetes, and sexual dysfunction, whereas the MC-4R antagonists are useful for the treatment of disorders and diseases such as cancer cachexia, muscle wasting, anorexia, anxiety and depression. All diseases and disorders where the regulation of the MC-4R is involved can be treated with the compounds of the invention.

Description

    FIELD OF THE INVENTION
  • The present invention relates to novel substituted piperidine and piperazine derivatives as melanocortin-4 receptor modulators. Depending on the structure and the stereochemistry the compounds of the invention are either selective agonists or selective antagonists of the human melanocortin-4 receptor (MC-4R). The agonists can be used for the treatment of disorders and diseases such as obesity, diabetes and sexual dysfunction, whereas the antagonists are useful for the treatment of disorders and diseases such as cancer cachexia, muscle wasting, anorexia, anxiety and depression. Generally all diseases and disorders where the regulation of the MC-4R is involved can be treated with the compounds of the invention.
  • BACKGROUND OF THE INVENTION
  • Melanocortins (MCs) stem from pro-opiomelanocortin (POMC) via proteolytic cleavage.
  • These peptides, adrenocorticotropic hormone (ACTH), α-melanocyte-stimulating hormone (α-MSH), β-MSH and γ-MSH, range in size from 12 to 39 amino acids. The most important endogenous agonist for central MC-4R activation appears to be the tridecapeptide α-MSH. Among MCs, it was reported that α-MSH acts as a neurotransmitter or neuromodulator in the brain. MC peptides, particularly α-MSH, have a wide range of effects on biological functions including feeding behavior, pigmentation and exocrine function. The biological effects of α-MSH are mediated by a sub-family of 7-transmembrane G-protein-coupled receptors, termed melanocortin receptors (MC-Rs). Activation of any of these MC-Rs results in stimulation of cAMP formation.
  • To date, five distinct types of receptor subtype for MC (MC-1R to MC-5R) have been identified and these are expressed in different tissues.
  • MC-1R was first found in melanocytes. Naturally occurring inactive variants of MC-1R in animals were shown to lead to alterations in pigmentation and a subsequent lighter coat color by controlling the conversion of phaeomelanin to eumelanin through the control of tyrosinase. From these and other studies, it is evident that MC-1R is an important regulator of melanin production and coat color in animals and skin color in humans.
  • The MC-2R is expressed in the adrenal gland representing the ACTH receptor. The MC-2R is not a receptor for α-MSH but is the receptor for the adrenocorticotropic hormone I (ACTH I).
  • The MC-3R is expressed in the brain (predominately located in the hypothalamus) and peripheral tissues like gut and placenta, and knock-out studies have revealed that the MC-3R may be responsible for alterations in feeding behavior, body weight and thermogenesis.
  • The MC-4R is primarily expressed in the brain. Overwhelming data support the role of MC-4R in energy homeostasis. Genetic knock-outs and pharmacologic manipulation of MC-4R in animals have shown that agonizing the MC-4R causes weight loss and antagonizing the MC-4R produces weight gain (A. Kask, et al., “Selective antagonist for the melanocortin-4 receptor (HS014) increases food intake in free-feeding rats,” Biochem. Biophys. Res. Commun., 245: 90-93 (1998)).
  • MC-5R is ubiquitously expressed in many peripheral tissues including white fat, placenta and a low level of expression is also observed in the brain. However its expression is greatest in exocrine glands. Genetic knock-out of this receptor in mice results in altered regulation of exocrine gland function, leading to changes in water repulsion and thermoregulation. MC-5R knockout mice also reveal reduced sebaceous gland lipid production (Chen et al., Cell, 91: 789-798 (1997)).
  • Attention has been focused on the study of MC-3R and MC-4R modulators and their use in treating body weight disorders, such as obesity and anorexia. However, evidence has shown that the MC peptides have potent physiological effects besides their role in regulating pigmentation, feeding behavior and exocrine function. In particular, α-MSH recently has been shown to induce a potent anti-inflammatory effect in both acute and chronic models of inflammation including inflammatory bowel-disease, renal ischemia/reperfusion injury and endotoxin-induced hepatitis. Administration of α-MSH in these models results in substantial reduction of inflammation-mediated issue damage, a significant decrease in leukocyte infiltration and a dramatic reduction in elevated levels of cytokines and other mediators to near baseline levels. Recent studies have demonstrated that the anti-inflammatory actions of α-MSH are mediated by MC-1R. The mechanism by which agonism of MC-1R results in an anti-inflammatory response is likely through inhibition of the pro-inflammatory transcription activator, NF-κB. NF-κB is a pivotal component of the pro-inflammatory cascade, and its activation is a central event in initiating many inflammatory diseases. Additionally, anti-inflammatory actions of α-MSH may be, in part, mediated by agonism of MC-3R and/or MC-5R.
  • A specific single MC-R that may be targeted for the control of obesity has not yet been identified, although evidence has been presented that MC-4R signaling is important in mediating feeding behavior (S. Q. Giraudo et al., “Feeding effects of hypothalamic injection of melanocortin-4 receptor ligands,” Brain Research, 80: 302-306 (1998)). Further evidence for the involvement of MC-Rs in obesity includes: a) the agouti (Avy) mouse which ectopically expresses an antagonist of the MC-1R. MC-3R and MC-4R is obese, indicating that blocking the action of these three MC-R's can lead to hyperphagia and metabolic disorders; 2) MC-4R knockout mice (D. Huszar et al., Cell, 88: 131-141 (1997)) recapitulate the phenotype of the agouti mouse and these mice are obese; 3) the cyclic heptapeptide melanotanin II (MT-II) (a non-selective MC-1R, -3R, -4R, and -5R agonist) injected intracerebroventricularly (ICV) in rodents, reduces food intake in several animal feeding models (NPY, ob/ob, agouti, fasted) while ICV injected SHU-9119 (MC-3R and 4R antagonist; MC-1R and -5R agonist) reverses this effect and can induce hyperphagia; 4) chronic intraperitoneal treatment of Zucker fatty rats with an α-NDP-MSH derivative (HP-228) has been reported to activate MC-1R, -3R, -4R, and -5R and to attenuate food intake and body weight gain over a 12 week period (I. Corcos et al., “HP-228 is a potent agonist of melanocortin receptor-4 and significantly attenuates obesity and diabetes in Zucker fatty rats,” Society for Neuroscience Abstracts, 23: 673 (1997)).
  • MC-4R appears to play a role in other physiological functions as well, namely controlling grooming behavior, erection and blood pressure. Erectile dysfunction denotes the medical condition of inability to achieve penile erection sufficient for successful intercourse. The term “impotence” is often employed to describe this prevalent condition. Synthetic melanocortin receptor agonists have been found to initiate erections in men with psychogenic erectile dysfunction (H. Wessells et al., “Synthetic Melanotropic Peptide Initiates Erections in Men With Psychogenic Erectile Dysfunction: Double-Blind, Placebo Controlled Crossover Study,” J. Urol., 160: 389-393, 1998). Activation of melanocortin receptors of the brain appears to cause normal simulation of sexual arousal. Evidence for the involvement of MC-R in male and/or female sexual dysfunction is detailed in WO/0074679.
  • Diabetes is a disease in which a mammal's ability to regulate glucose levels in the blood is impaired because the mammal has a reduced ability to convert glucose to glycogen for storage in muscle and liver cells. In Type I diabetes, this reduced ability to store glucose is caused by reduced insulin production. “Type II diabetes” or “Non-Insulin Dependent Diabetes Mellitus” (NIDDM) is the form of diabetes which is due to a profound resistance to insulin stimulating or regulatory effect on glucose and lipid metabolism in the main insulin-sensitive tissues, muscle, liver and adipose tissue. This resistance to insulin responsiveness results in insufficient insulin activation of glucose uptake, oxidation and storage in muscle, and inadequate insulin repression of lipolysis in adipose tissue and of glucose production and secretion in liver. When these cells become desensitized to insulin, the body tries to compensate by producing abnormally high levels of insulin and hyperinsulemia results. Hyperinsulemia is associated with hypertension and elevated body weight. Since insulin is involved in promoting the cellular uptake of glucose, amino acids and triglycerides from the blood by insulin sensitive cells, insulin insensitivity can result in elevated levels of triglycerides and LDL which are risk factors in cardiovascular diseases. The constellation of symptoms which includes hyperinsulemia combined with hypertension, elevated body weight, elevated triglycerides and elevated LDL, is known as Syndrome X. MC-4R agonists might be useful in the treatment of NIDDM and Syndrome X.
  • Among MC receptor subtypes, the MC-4 receptor is also of interest in terms of the relationship to stress and the regulation of emotional behavior, as based on the following findings. Stress initiates a complex cascade of responses that include endocrine, biochemical and behavioral events. Many of these responses are initiated by release of corticotropin-releasing factor (CRF) (Owen M J and Nemeroff C B (1991) Physiology and pharmacology of corticotrophin releasing factor. Pharmacol Rev 43:425-473). In addition to activation of the brain CRF system, there are several lines of evidence that melanocortins (MCs), which stem from proopiomelanocortin by enzymatc processing, mediate important behavioral and biochemical responses to stress and, consequently, stress-induced disorders like anxiety and depression (Anxiolytic-Like and Antidepressant-Like Activities of MCL0129 (1-[(S)-2-(4-Fluorophenyl)-2-(4-isopropylpiperadin-1-yl)ethyl]-4-[4-(2-methoxynaphthalen-1-yl)butyl]piperazine), a Novel and Potent Nonpeptide Antagonist of the Melanocortin-4 Receptor; Shigeyuki Chaki et al, J. Pharm. Exp. Ther. (2003)304(2), 818-26).
  • Chronic diseases, such as malignant tumors or infections, are frequently associated with cachexia resulting from a combination of a decrease in appetite and a loss of lean body mass. Extensive loss of lean body mass is often triggered by an inflammatory process and is usually associated with increased plasma levels of cytokines (e.g. TNF-α), which increase the production of α-MSH in the brain. Activation of MC-4 receptors in the hypothalamus by α-MSH reduces appetite and increases energy expenditure. Experimental evidence in tumor bearing mice suggests that cachexia can be prevented or reversed by genetic MC-4 receptor knockout or MC-4 receptor blockade. The increased body weight in the treated mice is attributable to a larger amount of lean body mass, which mainly consists of skeletal muscle (Marks D. L. et al. Role of the central melanocortin system in cachexia. Cancer Res. (2001) 61: 1432-1438).
  • WO03009847A and WO03009850A describe phenylpiperazinyl-phenylalanine derivatives for the treatment of obesity. Most of the compounds in both patents contain a N-(2-piperidin-4-yl-phenyl)-alkyl, benzyl or aryl sulfonamide group and N-(2-piperazin-4-yl-phenyl)-alkyl, benzyl or aryl sulfonamide group, respectively. These compounds have in common the substituted phenylalanine moiety which is acylated with amino acids, carboxylic acids and sulfonyl chlorides. In some cases the phenylalanine moiety is also alkylated or uraes and urethanes are introduced. Biological data (e.g. binding IC50 or functional activity) are not provided.
  • WO02070511A describes phenylpiperazinyl-phenylalanine amides, phenylpiperidinyl-phenylalanine amides and cyclohexyl-phenylalanine amides as modulators of melanocortin receptors 1 and 4. The phenylalanine amino group is in the most cases acylated with a second amino acid. For amino acids with a basic side chain the amino group can be acylated. Biological data for the compounds are not given.
  • WO02059108A describes melanocortin receptor agonists. The agonists consist of substituted phenylpiperazines which are first acylated with phenylalanines and then with amino acids. Biological data are not provided.
  • WO0074679A describes substituted piperidines as melanocortin-4 receptor agonists. The piperidines are acylated with different substituted phenylalanines, e.g. D-p-chlorophenylalanine, which are subsequently acylated with other amino acids, in particular 1,2,3,4-tetrahydro-isoquinoline-3-carboxylic acid. Example 2 of this patent application binds to the human MC-4-receptor with an IC50 of 0.92 nM. The compound is acting as an agonist with an EC50 of 2.1 nM (96% activation).
  • WO9964002A describes spiroindane derivatives as melanocortin receptor agonists. The spiroindanes are acylated with phenylalanine, in particular p-chlorophenylalanine, which is then acylated with unsubstituted and substituted piperazine-2-carboxylic acid, morpholine-3-carboxylic acid and thiomorpholine-3-carboxylic acid, respectively. No biological data is given.
  • WO0170337A describes spiroindane derivatives as melanocortin receptor agonists. The spiroindanes are acylated with phenylalanine, in particular p-chlorophenylalanine, which is then acylated with unsubstituted and substituted 1,2,3,4-tetrahydro-isoquinoline-3-carboxylic acid. No biological data is given.
  • WO0191752A decribes melanocortin receptor agonists. Three examples are described consisting of 3a-benzyl-2-methyl-2,3a,4,5,6,7-hexahydro-pyrazolo[4,3-c]pyridin-3-one which is first acylated with Boc-D-4-chlorophenylalanine following a second acylation step using 1-amino-1,2,3,4-tetrahydro-naphthalene-2-carboxylic acid and 1,2,3,4-tetrahydro-isoquinoline-3-carboxylic acid, respectively. Biological data for the examples is not provided.
  • In view of the unresolved deficiencies in treatment of various diseases and disorders as discussed above, it is an object of the present invention to provide novel substituted piperidine and piperazine derivatives with improved ability to cross the blood brain barrier, which are useful as melanocortin-4 receptor modulators to treat cancer cachexia, muscle wasting, anorexia, anxiety, depression, obesity, diabetes, sexual dysfunction and other diseases with MC-4R involvement.
  • SUMMARY OF THE INVENTION
  • The present invention relates to novel substituted piperidine and piperazine derivatives of structural formula (I),
    Figure US20060241123A1-20061026-C00001

    wherein the variables A, R1, Ar, m and n have the meaning as defined below.
  • The present invention also relates to novel substituted piperidine and piperazine derivatives of structural formula (II),
    Figure US20060241123A1-20061026-C00002

    wherein the variables A, R1, Ar, m and n have a different meaning than in structural formula (I) and are also defined below.
  • The piperidine and piperazine derivatives of structural formulas (I) and (II) are effective as melanocortin receptor modulators and are particularly effective as selective melanocortin-4 receptor (MC-4R) modulators. They are Thereforee useful for the treatment of disorders where the activation or inactivation of the MC-4R are involved. Agonists can be used for the treatment of disorders and diseases such as obesity, diabetes and sexual dysfunction, whereas the antagonists are useful for the treatment of disorders and diseases such as cancer cachexia, muscle wasting, anorexia, anxiety and depression.
  • The present invention also relates to pharmaceutical compositions comprising the compounds of the present invention and a pharmaceutically acceptable carrier.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to novel substituted piperidine and piperazine derivatives useful as melanocortin receptor modulators, in particular, selective MC-4R agonists and MC-4R antagonists.
  • The compounds of the present invention are represented by structural formula (I).
    Figure US20060241123A1-20061026-C00003

    or a pharmaceutically acceptable salt or a solvate thereof, wherein
    Ar is:
      • aryl or heteroaryl which may both be substituted;
        R1 is:
        Figure US20060241123A1-20061026-C00004

        A is:
        Figure US20060241123A1-20061026-C00005

        R2 is independently:
      • hydrogen,
      • halo,
      • alkyl,
      • haloalkyl,
      • hydroxy,
      • alkoxy,
      • S-alkyl,
      • SO2-alkyl,
      • O-alkenyl,
      • S-alkenyl,
      • NR14C(O)R14.
      • NR14SO2R14,
      • N(R14)2,
      • (D)-cycloalkyl,
      • (D)-aryl,
      • (D)-heteroaryl,
      • (D)-heterocyclyl (wherein heterocyclyl excludes a heterocyclyl containing a single nitrogen), and
      • wherein aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl are substituted or unsubstituted, and two adjacent R2 may form a 4- to 7-membered ring;
        R4 and R5 are each independently:
      • hydrogen,
      • alkyl,
      • (D)-cycloalkyl or
      • R4 and R5 together with the nitrogen to which they are attached form a 5- to 8-membered ring,
      • wherein alkyl and cycloalkyl are unsubstituted or substituted;
        R8 is independently:
      • hydrogen,
      • alkyl,
      • (D)-aryl or
      • (D)-cycloalkyl;
        R9 is independently:
      • hydrogen,
      • alkyl,
      • (D)-aryl,
      • (D)-heteroaryl or
      • (D)-cycloalkyl;
        R10 is independently:
      • R9,
      • (D)-heterocyclyl,
      • (D)-N(Y)2,
      • (D)-NH-heteroaryl or
      • (D)-NH-heterocyclyl,
      • wherein aryl and heteroaryl, alkyl, D, cycloalkyl and heterocyclyl are substituted or unsubstituted, or
      • two R10 groups together with the atoms to which they are attached form a 5- to 8-membered mono- or bi-cyclic ring system;
        R11 is:
      • hydrogen,
      • halo,
      • alkyl,
      • alkoxy,
      • C≡N,
      • CF3 or
      • OCF3;
        R12 is independently:
      • hydrogen,
      • hydroxy,
      • cyano,
      • nitro,
      • halo,
      • alkyl,
      • alkoxy,
      • haloalkyl,
      • (D)-C(O)R14,
      • (D)-C(O)OR14,
      • (D)-C(O)SR14,
      • (D)-C(O)-heteroaryl,
      • (D)-C(O)-heterocyclyl,
      • (D)-C(O)N(R14)2,
      • (D)-N(R14)2,
      • (D)-NR14COR14,
      • (D)-NR14CON(R14)2.
      • (D)-NR14C(O)OR14,
      • (D)-NR14C(R14)═N(R14),
      • (D)-NR14C(═NR14)N(R14)2,
      • (D)-NR14SO2R14,
      • (D)-NR14SO2N(R14)2,
      • (D)-NR14(D)-heterocyclyl,
      • (D)-NR14(D)-heteroaryl,
      • (D)-OR14,
      • OSO2R14,
      • (D)[O]q(cycloalkyl),
      • (D)[O]q(D)aryl,
      • (D)[O]q(D)-heteroaryl,
      • (D)[O]q(D)-heterocyclyl (wherein heterocyclyl excludes a heterocyclyl containing a single nitrogen when q=1),
      • (D)-SR14,
      • (D)-SOR14,
      • (D)-SO2R14 or
      • (D)-SO2N(R14)2,
      • wherein alkyl, alkoxy, cycloalkyl, aryl, heterocyclyl and heteroaryl are substituted or unsubstituted;
        R14 is independently:
      • hydrogen,
      • alkyl,
      • haloalkyl,
      • (D)-cycloalkyl,
      • (D)-phenyl,
      • (D)-naphthyl,
      • (D)-heteroaryl,
      • (D)-heterocyclyl (wherein heterocyclyl excludes a heterocyclyl containing a single nitrogen), and
      • wherein phenyl, naphthyl, heteroaryl, heterocyclyl, alkyl or cycloalkyl is substituted or unsubstituted;
        X is:
      • alkyl,
      • (D)-cycloalkyl,
      • (D)-aryl,
      • (D)-heteroaryl,
      • (D)-heterocyclyl,
      • (D)-C≡N,
      • (D)-CON(R9R9),
      • (D)-CO2R9,
      • (D)-COR9,
      • (D)-NR9C(O)R9,
      • (D)-NR9CO2R9,
      • (D)-NR9C(O)N(R9)2,
      • (D)-NR9SO2R9,
      • (D)-S(O)pR9,
      • (D)-SO2N(R9)(R9),
      • (D)-OR9,
      • (D)-OC(O)R9,
      • (D)-OC(O)OR9,
      • (D)-OC(O)N(R9)2,
      • (D)-N(R9)(R9) or
      • (D)-NR9SO2N(R9)(R9),
      • wherein aryl, heteroaryl, alkyl, D, cycloalkyl and heterocyclyl are unsubstituted or substituted;
        Y is:
      • hydrogen,
      • alkyl,
      • (D)-cycloalkyl,
      • (D)-aryl,
      • (D)-heterocyclyl or
      • (D)-heteroaryl,
      • wherein aryl, heteroaryl, alkyl, D and cycloalkyl are unsubstituted or substituted;
        Cy is benzene, pyridine or cyclohexane;
        D is a bond or alkylen;
        E is CHCO2Y, CHC(O)N(Y)2, NSO2R10, CHN(Y)COR10, CHN(Y)SO2R10, CHCH2OY or CHCH2heteroaryl;
        G is D, CH-alkyl, O, C═O or SO2, with the proviso that when G is O, the ring atom E is carbon;
        J is N or CH;
        T is O or NR4;
        n is 0-2;
        m is 0-2;
        o is 0-3;
        p is 0-2;
        q is 0 or 1;
        r is 1 or 2.
  • In preferred embodiments of formula I, the variants have the following meanings:
  • Ar is as defined above, and is preferably aryl, more preferably phenyl or naphthyl. If aryl or heteroaryl are substituted, it is preferably substituted with one to three, more preferably one or two, most preferably one, substituents. The substituents are preferably independently selected from the group consisting of cyano, nitro, perfluoroalkoxy, halo, alkyl, (D)-cycloalkyl, alkoxy and haloalkyl, more preferably perfluoroalkoxy, halo, alkyl, alkoxy or haloalkyl, even more preferably halo, alkyl, alkoxy and/or haloalkyl, in particular halo.
  • Most preferably, Ar is phenyl or naphthyl which both, preferably phenyl, may be substituted with one to three, in particular one, halo, e.g. Cl. The substitution can be in any position, preferably in the 4-position.
    R1 is as defined above preferably:
    Figure US20060241123A1-20061026-C00006

    A is:
    Figure US20060241123A1-20061026-C00007

    R2 is as defined above. If aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl are substituted, they are preferably independently substituted with one to three, more preferably one substituent selected from the group consisting of oxo, halo, alkyl, N(R6)2, OR6, SR6 and/or CO2R6.
  • Preferably, R2 is hydrogen, hydroxy, halo, alkyl, alkoxy, S-alkyl, SO2-alkyl, O-alkenyl, S-alkenyl, haloalkyl or (D)-cycloalkyl, more preferably hydrogen, hydroxy, alkoxy, S-alkyl, SO2-alkyl, O-alkenyl, S-alkenyl, halo or alkyl, e.g. methyl, ethyl, n-propyl, isopropyl. In one embodiment, R2 is hydrogen, halo, alkyl, haloalkyl, alkoxy, (D)-cycloalkyl, (D)-aryl, (D)-heteroaryl, (D)-heterocyclyl (wherein heterocyclyl excludes a heterocyclyl containing a single nitrogen), and wherein aryl, heteroaryl, heterocyclyl, alkyl and cycloalkyl are substituted or unsubstituted; preferably hydrogen, hydroxy, halo, alkyl, alkoxy, haloalkyl or (D)-cycloalkyl, more preferably hydrogen, alkoxy, halo or alkyl.
  • R4 and R5 are each independently as defined above. When R4 and R5 form a ring, said ring may contain an additional heteroatom preferably selected from O, S and NR6 in the ring. Moreover, if alkyl and cycloalkyl are substituted, they are preferably substituted with one to three, more preferably one or two groups independently selected from R7 and oxo.
  • R4 and R5 are each independently preferably selected from the group consisting of hydrogen, alkyl or cycloalkyl; or R4 and R5 together with the nitrogen to which they are attached form a 5- to 7-membered ring. More preferably R4 and R5 are each independently selected from the group consisting of hydrogen or alkyl, or R4 and R5 together with the nitrogen to which they are attached form a 5- to 6-membered ring optionally containing an additional oxygen atom.
  • R6 is independently hydrogen, alkyl, C(O) alkyl, (D)-aryl or (D)-cycloalkyl, preferably hydrogen, alkyl, C(O) alkyl, (D)-aryl or (D)-cycloalkyl, most preferably hydrogen.
  • R7 is alkyl, (D)-aryl, (D)-cycloalkyl, (D)-heteroaryl, halo, OR8, NHSO2R8, N(R8)2, C≡N, CO2R4, C(R8)(R8)N(R8)2, nitro, SO2N(R8)2, S(O)pR8, CF3 or OCF3, preferably hydrogen, alkyl, (D)-aryl or (D)-cycloalkyl, more preferably hydrogen.
  • R8 is as defined above, preferably hydrogen, alkyl or (D)-aryl, more preferably alkyl or (D)-aryl.
  • R9 is as defined above, preferably hydrogen, alkyl and (D)-cycloalkyl, more preferably alkyl, most preferably methyl, ethyl or tert-butyl.
  • R10 is as defined above. If aryl and heteroaryl are substituted, they are preferably independently substituted with one to three groups selected from R7. Moreover, if alkyl, D, cycloalkyl and heterocyclyl are substituted, they are preferably substituted with one to four groups independently selected from R7 and oxo. If two R10 groups together with the atoms to which they are attached form a 5- to 8-membered mono- or bi-cyclic ring system, said ring may contain an additional heteroatom preferably selected from O, S, NR8, NBoc and NZ.
  • Preferably, R10 is R9.
  • R11 is defined as above, preferably hydrogen, halo, alkyl, alkoxy or C≡N, more preferably hydrogen, halo or C1-C4-alkyl, most preferably hydrogen.
  • R12 is as defined above. Moreover, if alkyl, alkoxy, cycloalkyl, aryl, heterocyclyl and heteroaryl are substituted, they are preferably substituted with 1 to 5, more preferably 1 to 3, most preferably 1 or 2 substituents independently selected from R13.
  • Preferably, R12 is hydrogen, hydroxy, cyano, nitro, halo, alkyl, alkoxy, haloalkyl, (D)-C(O)-heterocyclyl, (D)-C(O)N(R14)2, (D)-N(R14)2, (D)-NR14COR14, (D)-NR14CON(R14)2, (D)-NR14C(O)OR14, (D)-NR14C(R14)═N(R14), (D)-NR14C(═NR14)N(R14)2, (D)-NR14SO2R14 or (D)-NR14SO2N(R14)2, wherein alkyl or alkoxy are substituted or unsubstituted with one to five, preferably one to three, substituents selected from R13. More preferably, R12 is cyano, nitro, halo, alkyl, D)-C(O)-heterocyclyl, (D)-N(R14)2 (D)-NR14COR14, (D)-NR14CON(R14)2, (D)-NR14C(O)OR14 or (D)-NR14SO2R14. Most preferably, R12 is (D)-C(O)-heterocyclyl, (D)-C(O)N(R14)2. Halo is preferably F, Cl or Br. R12 can be on any position of the ring, preferably in the 1-position. In one embodiment, R12 is hydrogen, hydroxy, cyano, nitro, halo, alkyl, alkoxy, haloalkyl, (D)-N(R14)2, (D)-NR14COR14, (D)-NR14CON(R14)2, (D)-NR14C(O)OR14, (D)-NR14C(R14)═N(R14), (D)-NR14C(═NR14)N(R14)2, (D)-NR14SO2R14 or (D)-NR14SO2N(R14)2, wherein alkyl or alkoxy are substituted or unsubstituted with one to five, preferably one to three, substituents selected from R13. More preferably, R12 is cyano, nitro, halo, alkyl, (D)-N(R14)2 (D)-NR14COR14, (D)-NR14CON(R14)2, (D)-NR14C(O)OR14 or (D)-NR14SO2R14.
  • R13 is independently hydrogen, halo, oxo, N(R15)2, alkyl, (D)-cycloalkyl, haloalkyl, alkoxy, heteroaryl, hydroxy or heterocyclyl, wherein heterocyclyl excludes a heterocyclyl containing a single nitrogen, phenyl, (D)-COR14, (D)-C(O)OR14, (D)-OR14, (D)-OCOR14, (D)-OCO2R14, (D)-SR14, (D)-SOR14 or (D)-SO2R14, wherein aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl is substituted or unsubstituted. If aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl are substituted, they are preferably substituted with one to three, preferably one or two, substituents selected from the group consisting of oxo, alkyl, N(R15)2, OR15, SR15 and/or CO2R15.
  • Preferably, R13 is hydrogen, halo, alkyl, (D)-cycloalkyl, alkoxy or phenyl, more preferably R13 is hydrogen, halo, alkyl, alkoxy or phenyl.
  • R14 is as defined above. If aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl are substituted, they are preferably substituted with one to three, more preferably one or two, substituents selected from the group consisting of oxo, halo, alkyl, N(R15)2, OR15, SR15 and/or CO2R15.
  • Preferably, R14 is hydrogen, haloalkyl, alkyl, (D)-cycloalkyl or phenyl, more preferably R14 is hydrogen, haloalkyl, alkyl or phenyl.
  • R15 is independently hydrogen, alkyl, C(O)alkyl, aryl or cycloalkyl, preferably hydrogen, alkyl or cycloalkyl, in particular, hydrogen.
  • X is as defined above. If aryl and heteroaryl are substituted, they are preferably substituted with one to three, more preferably one or two, groups selected from R7. Moreover, if alkyl, D, cycloalkyl and heterocyclyl are substituted, they are preferably substituted with one to four groups independently selected from R7 and oxo.
  • Preferably, X is alkyl, (D)-cycloalkyl, (D)-aryl, (D)-heteroaryl, (D)-heterocyclyl, (D)-NHC(O)R9, (D)-CO2R9 or (D)-CON(R9R9), more preferably from alkyl, (D)-cycloalkyl, (D)-heterocyclyl, (D)-NHC(O)R9 or (D)-CON(R9R9), most preferably from C1-C4-alkyl, C5-C7-cycloalkyl, (D)-CON(R9R9) and N-containing heterocyclyl, in particular triazolyl and tetrazolyl.
  • Y is as defined above. If aryl and heteroaryl are substituted, they are preferably substituted with one to three, more preferably one or two, groups selected from R7. Moreover if alkyl, D and cycloalkyl are substituted, they are preferably substituted with one to three groups selected from R7 and oxo.
  • Preferably, Y is hydrogen, alkyl, (D)-cycloalkyl, (D)-aryl, (D)-heteroaryl or (D)-heterocyclyl, more preferably hydrogen, alkyl, (D)-cycloalkyl or (D)-heterocyclyl, most preferably hydrogen, C1-C4-alkyl or C5-C7-cycloalkyl, in particular cyclohexyl.
  • Cy is as defined above, preferably benzene or pyridine, more preferably benzene.
  • D is as defined above, preferably a bond or C1-C4-alkylene, more preferably a bond or CH2.
  • E is as defined above, preferably NSO2R10, CHN(Y)COR10 or CHN(Y)SO2R10, more preferably NSO2R10.
  • G is as defined above, preferably D or CH-alkyl, more preferably D, in particular CH2.
  • J is N or CH;
  • T is O or NR4, preferably 0; in one embodiment, T is NR4;
  • n is 0, 1 or 2, preferably 0 or 1, more preferably 0;
  • m is 0, 1 or 2, preferably 0 or 1, more preferably 0;
  • o is 0, 1, 2 or 3, preferably 0, 1 or 2, more preferably 0 or 1;
  • p is 0, 1 or 2;
  • q is 0 or 1;
  • r is 1 or 2, preferably 1.
  • Compounds of the present invention are also represented by structural formula (II),
    Figure US20060241123A1-20061026-C00008

    or a pharmaceutically acceptable salt or a solvate thereof, wherein
    Ar is:
      • aryl or heteroaryl, which may both be substituted;
        R1 is:
        Figure US20060241123A1-20061026-C00009

        A is:
        Figure US20060241123A1-20061026-C00010

        R2 is independently:
      • hydrogen,
      • halo,
      • alkyl,
      • haloalkyl,
      • alkoxy,
      • (D)-cycloalkyl,
      • (D)-aryl,
      • (D)-heteroaryl,
      • (D)-heterocyclyl (wherein heterocyclyl excludes a heterocyclyl containing a single nitrogen), and
      • wherein aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl are substituted or unsubstituted.
        R3 is independently:
      • hydrogen,
      • alkyl,
      • SO2alkyl,
      • SO2aryl,
      • C(O)alkyl,
      • (D)-aryl or
      • cycloalkyl;
        R4 is independently:
      • hydrogen,
      • alkyl,
      • (D)-aryl,
      • (D)-heteroaryl,
      • (D)-N(R6)2,
      • (D)-NR6C(O)alkyl,
      • (D)-NR6SO2alkyl,
      • (D)-SO2N(R6)2,
      • (D)-(O)valkyl,
      • (D)-(O)v(D)NR6COR6,
      • (D)-(O)v(D)NR7SO2R7,
      • (D)-(O)v-heterocyclyl or
      • (D)-(O)v(alkyl)-heterocyclyl;
        R5 is independently:
      • hydrogen,
      • alkyl,
      • (D)-phenyl,
      • C(O)alkyl,
      • C(O)phenyl,
      • SO2-alkyl or
      • SO2-phenyl;
        R6 and R7 are each independently:
      • hydrogen,
      • alkyl,
      • cycloalkyl, or
      • R6 and R7 together with the nitrogen to which they are attached form a 5- to 8-membered ring,
      • wherein alkyl and cycloalkyl are unsubstituted or substituted;
        R9 is independently:
      • hydrogen,
      • alkyl,
      • (D)-aryl or
      • cycloalkyl;
        R10 is hydrogen or alkyl;
        R11 is independently:
      • hydrogen,
      • alkyl,
      • (D)-aryl,
      • (D)-heteroaryl or
      • (D)-cycloalkyl;
        R12 is independently:
      • (D)-heterocyclyl,
      • (D)-N(Y)2,
      • (D)-NH-heteroaryl or
      • (D)-NH-heterocyclyl,
      • wherein aryl, heteroaryl, alkyl, D, cycloalkyl and heterocyclyl are unsubstituted or substituted, or
      • two R12 groups together with the atoms to which they are attached form a 5- to 8-membered mono- or bicyclic ring system;
        R13 is:
      • hydrogen,
      • halo,
      • alkyl,
      • alkoxy,
      • C≡N,
      • CF3 or
      • OCF3;
        R14 is independently:
      • hydrogen,
      • hydroxy,
      • cyano,
      • nitro,
      • halo,
      • alkyl,
      • alkoxy,
      • haloalkyl,
      • (D)-C(O)R16,
      • (D)-C(O)OR16,
      • (D)-C(O)SR16,
      • (D)-C(O)-heteroaryl,
      • (D)-C(O)-heterocyclyl,
      • (D)-C(O)N(R16)2.
      • (D)-N(R16)2,
      • (D)-NR16COR16,
      • (D)-NR16CON(R16)2,
      • (D)-NR16C(O)OR16,
      • (D)-NR16C(R16)═N(R16),
      • (D)-NR16C(═NR16)N(R16)2,
      • (D)-NR16SO2R16,
      • (D)-NR16SO2N(R16)2,
      • (D)-NR16(D)-heterocyclyl,
      • (D)-NR16(D)-heteroaryl,
      • (D)-OR16,
      • OSO2R16,
      • (D)-[O]q(cycloalkyl),
      • (D)-[O]q(D)aryl,
      • (D)-[O]q(D)-heteroaryl,
      • (D)-[O]q(D)-heterocyclyl (wherein heterocyclyl excludes a heterocyclyl containing a single nitrogen when q=1),
      • (D)-SR16,
      • (D)-SOR16,
      • (D)-SO2R16 or
      • (D)-SO2N(R16)2,
      • wherein alkyl, alkoxy, cycloalkyl, aryl, heterocyclyl and heteroaryl are substituted or unsubstituted;
        R16 is independently:
      • hydrogen,
      • alkyl,
      • haloalkyl,
      • (D)-cycloalkyl,
      • (D)-aryl,
      • (D)-heteroaryl,
      • (D)-heterocyclyl (wherein heterocyclyl excludes a heterocyclyl containing a single nitrogen), and
      • wherein aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl is substituted or unsubstituted;
        Cy is:
      • aryl,
      • heteroaryl,
      • heterocyclyl or
      • carbocyclyl;
        X is:
      • alkyl,
      • (D)-cycloalkyl,
      • (D)-aryl,
      • (D)-heteroaryl,
      • (D)-heterocyclyl,
      • (D)-C≡N,
      • (D)-CON(R11R11),
      • (D)-CO2R11,
      • (D)-COR11,
      • (D)-NR11C(O)R11,
      • (D)-NR11CO2R11,
      • (D)-NR11C(O)N(R11)2,
      • (D)-NR11SO2R11,
      • (D)-S(O)pR11,
      • (D)-SO2N(R11)(R11),
      • (D)-OR11,
      • (D)-OC(O)R11,
      • (D)-OC(O)OR11,
      • (D)-OC(O)N(R11)2,
      • (D)-N(R11)(R11) or
      • (D)-NR11SO2N(R11)(R11),
      • wherein aryl, heteroaryl, alkyl, D, cycloalkyl and heterocyclyl are unsubstituted or substituted;
        Y is:
      • hydrogen,
      • alkyl,
      • (D)-cycloalkyl,
      • (D)-aryl,
      • (D)-heterocyclyl or
      • (D)-heteroaryl,
      • wherein aryl, heteroaryl, alkyl, D and cycloalkyl are unsubstituted or substituted;
        Cy′ is benzene, pyridine or cyclohexane;
        D is a bond or alkylene;
        E is CHCO2Y, CHC(O)N(Y)2, NSO2R10, CHN(Y)COR12, CHN(Y)SO2R12, CHCH2OY or CHCH2heteroaryl;
        G is D, CH-alkyl, O, C═O or SO2, with the proviso that when G is O, the ring atom E is carbon;
        J is N or CH;
        L is O, S or NR5;
        M is bond, O, S(O)u, NR5 or CH2;
        n is 0-2, unless m is 0, then n is 1 or 2;
        m is 0-2, unless n is 0, then m is 1 or 2;
        o is 0-3;
        p is 0-2;
        q is 0 or 1;
        r is 1 or 2;
        s is 0-5;
        u is 0-2;
        v is 0 or 1.
  • In preferred embodiments of formula II, the variants have the following meanings:
  • Ar is as defined above, and is preferably aryl, more preferably phenyl or naphthyl. If aryl or heteroaryl are substituted, it is preferably substituted with one to three, more preferably one or two, most preferably one, substituents. The substituents are preferably independently selected from the group consisting of: cyano, nitro, perfluoroalkoxy, halo, alkyl, (D)-cycloalkyl, alkoxy and haloalkyl, more preferably cyano, perfluoroalkoxy, halo, alkyl, (D)-cycloalkyl, alkoxy or haloalkyl, even more preferably halo, alkyl, alkoxy and/or haloalkyl, in particular halo.
  • Most preferably, Ar is phenyl or naphthyl which both, preferably phenyl, may be substituted with one to three, in particular one, halo, e.g. Cl. The substitution can be in any position, preferably in the 4-position.
    R1 is as defined above, preferably:
    Figure US20060241123A1-20061026-C00011
  • More preferably, R1 is:
    Figure US20060241123A1-20061026-C00012

    A is as defined above.
    R2 is as defined above. If aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl are substituted, they are preferably substituted with one to three substituents, more preferably one, selected from the group consisting of oxo, alkyl, N(R3)2, OR3, SR3 and/or CO2R3.
  • Preferably, R2 is hydrogen, halo, alkyl, haloalkyl, alkoxy or (D)-cycloalkyl, more preferably hydrogen, halo or alkyl, e.g. methyl, ethyl, n-propyl, iso-propyl, most preferably hydrogen.
  • R3 is as defined above, preferably hydrogen or alkyl, e.g. methyl, ethyl, n-propyl, iso-propyl, more preferably hydrogen.
  • R4 is as defined above, preferably hydrogen, (D)-aryl, (D)-heteroaryl, (D)-N(R6)2, (D)-NR6C(O)alkyl, (D)-NR6SO2alkyl or alkyl, e.g. methyl, ethyl, n-propyl, iso-propyl, more preferably hydrogen.
  • R5 is as defined above, preferably hydrogen or alkyl, e.g. methyl, ethyl, n-propyl, iso-propyl, more preferably hydrogen.
  • R6 and R7 are each independently as defined above. When R6 and R7 form a ring, said ring may contain an additional heteroatom preferably selected from O, S and NR3 in the ring. Moreover, if alkyl and cycloalkyl are substituted, they are preferably substituted with one to three, more preferably one or two, groups independently selected from R8 and/or oxo.
  • R6 and R7 are each independently preferably selected from the group consisting of hydrogen, alkyl or cycloalkyl, or R6 and R7 together with the nitrogen to which they are attached form a 5- to 7-membered ring. More preferably R6 and R7 are each independently selected from the group consisting of hydrogen or alkyl, or R6 and R7 together with the nitrogen to which they are attached form a 5- to 6-membered ring optionally containing an additional oxygen atom.
  • R8 is alkyl, (D)-aryl, (D)-cycloalkyl, (D)-heteroaryl, halo, OR9, NHSO2R9, N(R9)2, C≡N, CO2R6, C(R9)(R9)N(R9)2, nitro, SO2N(R9)2, S(O)uR9, CF3 or OCF3. Preferably R8 is alkyl, OR9, (D)-aryl, (D)-cycloalkyl, (D)-heteroaryl or halo.
  • R9 is as defined above, preferably hydrogen, (D)-aryl or alkyl, e.g. methyl, ethyl, n-propyl, iso-propyl, more preferably hydrogen or (D)-aryl.
  • R10 is as defined above, preferably hydrogen or C1-C4 alkyl as defined below, more preferaby hydrogen, methyl or ethyl, most preferably hydrogen or methyl.
  • R11 is as defined above, preferably hydrogen or alkyl, more preferably hydrogen or C1-C6 alkyl as defined below, in particular C1-C5 alkyl, e.g. methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl and n-pentyl, most preferably hydrogen or tert-butyl.
  • R12 is as defined above. If aryl and heteroaryl are substituted, they are preferably substituted with one to three, more preferably one or two, groups selected from R7. Moreover, if alkyl, D, cycloalkyl and heterocyclyl are substituted, they are preferably substituted with one to four, more preferably one or two, groups independently selected from R8 and/or oxo. Furthermore, if two R12 groups together with the atoms to which they are attached form a 5- to 8-membered mono- or bi-cyclic ring system, said ring may contain an additional heteroatom preferably selected from O, S, NR9, NBoc and NZ.
  • Preferably, R12 is R11.
  • R13 is as defined above, preferably hydrogen, halo, alkyl, alkoxy and C≡N, more preferably hydrogen, methyl or ethyl, most preferably hydrogen or methyl.
  • R14 is as defined above. Moreover, if alkyl, alkoxy, cycloalkyl, aryl, heterocyclyl and heteroaryl are substituted, they are preferably substituted with 1 to 5, more preferably 1 to 3, most preferably 1 or 2, substituents independently selected from R15.
  • Preferably, R14 is hydrogen, hydroxy, cyano, nitro, halo, alkyl, alkoxy, haloalkyl, (D)-C(O)-heterocyclyl, (D)-C(O)N(R16)2, (D)-N(R16)2, (D)-NR16COR14, (D)-NR14CON(R14)2, (D)-NR14C(O)OR16, (D)-NR16C(R16)═N(R16), (D)-NR16C(═NR16)N(R16)2, (D)-NR16SO2R16 or (D)-NR16SO2N(R16)2, wherein alkyl or alkoxy are substituted or unsubstituted with one to five, preferably one to three, substituents selected from R15. More preferably, R14 is cyano, nitro, halo, alkyl, (D)-C(O)-heterocyclyl, (D)-C(O)N(R16)2, (D)-N(R16)2, (D)-NR16COR16, (D)-NR16CON(R16)2, (D)-NR16C(O)OR16 or (D)-NR16SO2R16. Most preferably, R16 is (D)-C(O)-heterocyclyl, (D)-C(O)N(R16)2 or NR16COR16. Halo is preferably F, Cl or Br. R14 can be on any position of the ring, preferably in the 1-position. In one embodiment, R14 is hydrogen, hydroxy, cyano, nitro, halo, alkyl, alkoxy, haloalkyl, (D)-N(R16)2, (D)-NR16COR14, (D)-NR14CON(R14)2, (D)-NR14C(O)OR16, (D)-NR16C(R16)═N(R16), (D)-NR16C(═NR16)N(R16)2, (D)-NR16SO2R16 or (D)-NR16SO2N(R16)2, wherein alkyl or alkoxy are substituted or unsubstituted with one to five, preferably one to three, substituents selected from R15. More preferably, R14 is cyano, nitro, halo, alkyl, (D)-N(R16)2, (D)-NR16COR16, (D)-NR16CON(R16)2, (D)-NR16C(O)OR16 or (D)-NR16SO2R16.
  • R15 is independently hydrogen, halo, oxo, N(R3)2, alkyl, (D)-cycloalkyl, alkoxy, haloalkyl, heteroaryl, hydroxy, heterocyclyl, wherein heterocyclyl excludes a heterocyclyl containing a single nitrogen, phenyl, (D)-COR16, (D)-C(O)OR16, (D)-R16, (D)-COR16, (D)-OCO2R16, (D)-SR16, (D)-SOR16 or (D)-SO2R16, wherein aryl, heteroaryl, heterocyclyl, alkyl and cycloalkyl are substituted or unsubstituted. If aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl are substituted, they are preferably substituted with one to three, more preferably one or two, substituents selected from the group consisting of oxo, alkyl, N(R16)2, OR16, SR16 and/or CO2R16.
  • Preferably, R15 is hydrogen, halo, alkyl, (D)-cycloalkyl, alkoxy or phenyl, more preferably R15 is hydrogen, halo, alkyl, alkoxy or phenyl.
  • R16 is as defined above. If aryl, heteroaryl, heterocyclyl, alkyl or cycloalkyl are substituted, they are preferably substituted with one to three, more preferably one or two, substituents selected from the group consisting of oxo, alkyl, N(R3)2, OR3, SR3 and/or CO2R3.
  • Preferably, R16 is hydrogen, halo, alkyl, (D)-cycloalkyl, alkoxy or phenyl, more preferably R16 is hydrogen, halo, alkyl, alkoxy or phenyl.
  • Cy is as defined above wherein heteroaryl and heterocyclyl are preferably 5- or 6-membered and carbocyclyl is preferably 5- or 7-membered. Preferably Cy is aryl, more preferably benzene.
  • X is as defined above. If aryl and heteroaryl are substituted, they are preferably substituted with one to three, more preferably one or two, groups selected from R8. Moreover, if alkyl, D, cycloalkyl and heterocyclyl are substituted, they are preferably substituted with one to four groups independently selected from R8 and/or oxo.
  • Preferably, X is alkyl, (D)-cycloalkyl, (D)-aryl, (D)-heteroaryl, (D)-heterocyclyl, (D)-NHC(O)R11, (D)-CO2R11 or (D)-CON(R11R11), more preferably alkyl, (D)-cycloalkyl, (D)-heterocyclyl, (D)-NHC(O)R1, or (D)-CON(R11R11), most preferably C1-C4-alkyl, C5-C7-cycloalkyl, (D)-CON(R11R11) and N-containing heterocyclyl, in particular triazolyl and tetrazolyl.
  • Y is as defined above. If aryl and heteroaryl are substituted, they are preferably substituted with one to three, more preferably one or two, groups selected from R8. Moreover, if alkyl, D and cycloalkyl are substituted, they are preferably substituted with one to three groups selected from R8 and/or oxo.
  • Preferably, Y is hydrogen, alkyl, (D)-cycloalkyl, (D)-aryl, (D)-heteroaryl or (D)-heterocyclyl, more preferably alkyl, (D)-cycloalkyl or (D)-heterocyclyl, most preferably hydrogen, C1-C4-alkyl and C5-C7-cycloalkyl, in particular cyclohexyl.
  • Cy′ is as defined above, preferably benzene or pyridine, more preferably benzene.
  • D is as defined above, preferably a bond or C1-C4 alkylene, more preferably a bond or CH2.
  • E is as defined above, preferably NSO2R10, CHN(Y)COR12 or CHN(Y)SO2R12, more preferably NSO2R10.
  • G is as defined above, preferably D or CH alkyl, more preferably D, in particular CH2.
  • J is as defined above;
  • L is as defined above, preferably NR5;
  • M is as defined above, preferably bond or CH2;
  • n is 0, 1 or 2, preferably 0 or 1, unless m is 0, then n is 1;
  • m is 0, 1 or 2, preferably 0 or 1, unless n is 0, then m is 1, more preferably n+m=1;
  • o is 0, 1, 2 or 3, preferably 0, 1 or 2, more preferably 1;
  • p is 0 or 1, preferably 0;
  • q is 0 or 1, preferably 0;
  • r is 1 or 2, preferably 1;
  • s is 0, 1, 2, 3, 4 or 5, preferably 0, 1, 2 or 3, more preferably 0 or 1, most preferably 0;
  • u is 0, 1 or 2;
  • v is 0 or 1.
  • In the above and the following, the employed terms have the meaning as described below:
  • Aryl is an aromatic mono- or polycyclic moiety with 6 to 20 carbon atoms which is preferably selected from phenyl, biphenyl, naphthyl, tetrahydronaphthyl, fluorenyl, indenyl or phenanthrenyl, more preferably from phenyl or naphthyl.
  • Heteroaryl is an aromatic moiety having 6 to 20 carbon atoms with at least one heterocycle and is preferably selected from thienyl, benzothienyl, naphthothienyl, furanyl, benzofuranyl, chromenyl, indolyl, isoindolyl, indazolyl, quinolyl, isoquinolyl, phthalazinyl, quinoxalinyl, cinnolinyl or quinazolinyl, more preferably from thienyl, furanyl, benzothienyl, benzofuranyl or indolyl.
  • Heterocyclyl is a saturated, unsaturated or aromatic ring containing at least one heteroatom selected from O, N and/or S and 1 to 6 carbon atoms and is preferably selected from azetidin-2-one-1-yl, pyrrolidin-2-one-1-yl, piperid-2-one-1-yl and azepan-2-one-1-yl, thienyl, furyl, piperidinyl, pyranyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, isothiazolyl or isoxazyl, more preferably from pyridyl, piperidinyl, imidazolyl or pyrazinyl.
  • Carbocyclyl is a monocyclic or polycyclic ring system of 3 to 20 carbon atoms which may be saturated, unsaturated or aromatic.
  • Alkyl is straight chain or branched alkyl having preferably 1 to 8 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, neopentyl, hexyl or heptyl, more preferably 1 to 4 carbon atoms.
  • Cycloalkyl is an alkyl ring having preferably 3 to 8 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl, more preferably 3 to 6 carbon atoms.
  • Alkenyl is straight chain or branched alkenyl having preferably 2 to 8 carbon atoms such as vinyl, allyl, methallyl, buten-2-yl, buten-3-yl, penten-2-yl, penten-3-yl, penten-4-yl, 3-methyl-but-3-enyl, 2-methyl-but-3-enyl, 1-methyl-but-3-enyl, hexenyl or heptenyl, more preferably 2 to 4 atoms.
  • Alkoxy is O-alkyl wherein alkyl is as defined above and has preferably 1 to 4 carbon atoms, preferably 1 or 3 carbon atoms.
  • Halo or halogen is a halogen atom preferably selected from F, Cl, Br and I, preferably F, Cl and Br.
  • Haloalkyl is an alkyl moiety as defined above having preferably 1 to 4 carbon atoms, more preferably 1 or 2 carbon atoms, wherein at least one, preferably 1 to 3 hydrogen atoms, have been replaced by a halogen atom. Preferred examples are —CF3, —CH2CF3 and —CF2CF3.
  • Therein, the alkylene moiety may be a straight chain or branched chain group. Said alkylene moiety preferably has 1 to 6 carbon atoms. Examples thereof include methylene, ethylene, n-propylene, n-butylene, n-pentylene, n-hexylene, iso-propylene, sec.-butylene, tert.-butylene, 1,1-dimethyl propylene, 1,2-dimethyl propylene, 2,2-dimethyl propylene, 1,1-dimethyl butylene, 1,2-dimethyl butylene, 1,3-dimethyl butylene, 2,2-dimethyl butylene, 2,3-dimethyl butylene, 3,3-dimethyl butylene, 1-ethyl butylene, 2-ethyl butylene, 3-ethyl butylene, 1-n-propyl propylene, 2-n-propyl propylene, 1-iso-propyl propylene, 2-iso-propyl propylene, 1-methyl pentylene, 2-methyl pentylene, 3-methyl pentylene and 4-methyl pentylene. More preferably, said alkylene moiety has 1 to 3 carbon atoms, such as methylene, ethylene, n-propylene and iso-propylene. Most preferred is methylene.
  • The compounds of structural formulas (I) and (II) are effective as melanocortin receptor modulators and are particularly effective as selective modulators of MC-4R. They are Thereforee useful for the treatment and/or prevention of disorders responsive to the activation and inactivation of MC-4R, such as cancer cachexia, muscle wasting, anorexia, anxiety, depression, obesity, diabetes, sexual dysfunction and other diseases with MC-4R involvement.
  • Optical Isomers—Diastereomers—Geometric Isomers—Tautomers
  • Compounds of structural formulas (I) and (II) contain one or more asymmetric centers and can occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. The present invention is meant to comprehend all such isomeric forms of the compounds of structural formulas (I) and (II).
  • Some of the compounds described herein may exist as tautomers, such as keto-enol tautomers. The individual tautomers, as well as mixtures thereof, are encompassed within the compounds of structural formulas (I) and (II).
  • Compounds of structural formulas (I) and (II) may be separated into their individual diastereoisomers by, for example, fractional crystallization from a suitable solvent, for example methanol or ethyl acetate or a mixture thereof, or via chiral chromatography using an optically active stationary phase. Absolute stereochemistry may be determined by X-ray crystallography of crystalline products or crystalline intermediates which are derivatized, if necessary, with a reagent containing an asymmetric center of known absolute configuration.
  • Alternatively, any stereoisomer of a compound of the general formulas (I) and (II) may be obtained by stereospecific synthesis using optically pure starting materials or reagents of known absolute configuration.
  • Salts
  • The term “pharmaceutically acceptable salts” refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids. Salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc and the like. Particularly preferred are the ammonium, calcium, lithium, magnesium, potassium and sodium salts. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N′-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like.
  • When the compound of the present invention is basic, salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids. Such acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, formic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, malonic, mucic, nitric, pamoic, pantothenic, phosphoric, propionic, succinic, sulfuric, tartaric, ptoluenesulfonic, trifluoroacetic acid and the like. Particularly preferred are citric, fumaric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric and tartaric acids.
  • It will be understood that, as used herein, references to the compounds of formulas (I) and (II) are meant to also include the pharmaceutically acceptable salts.
  • Utility
  • Compounds of formulas (I) and (II) are melanocortin receptor modulators and as such are useful in the treatment, control or prevention of diseases, disorders or conditions responsive to the activation or inactivation of one or more of the melanocortin receptors including, but not limited to, MC-1R, MC-2R, MC-3R, MC-4R or MC-5R. Such diseases, disorders or conditions include, but are not limited to, cancer cachexia, muscle wasting, anorexia, anxiety, depression, obesity (by reducing appetite, increasing metabolic rate, reducing fat intake or reducing carbohydrate craving), diabetes mellitus (by enhancing glucose tolerance, decreasing insulin resistance), hypertension, hyperlipidemia, osteoarthritis, cancer, gall bladder disease, sleep apnea, depression, anxiety, compulsion, neuroses, insomnia/sleep disorder, substance abuse, pain, male and female sexual dysfunction (including impotence, loss of libido and erectile dysfunction), fever, inflammation, immune-modulation, rheumatoid arthritis, skin tanning, acne and other skin disorders, neuroprotective and cognitive and memory enhancement including the treatment of Alzheimer's disease.
  • Some compounds encompassed by formulas (I) and (II) show highly selective affinity for the melanocortin-4 receptor relative to MC-1R, MC-2R, MC-3R and MC-5R, which makes them especially useful in the prevention and treatment of cancer cachexia, muscle wasting, anorexia, anxiety, depression, obesity, as well as male and/or female sexual dysfunction, including erectile dysfunction. “Male sexual dysfunction” includes impotence, loss of libido and erectile dysfunction. “Female sexual dysfunction” can be seen as resulting from multiple components including dysfunction in desire, sexual arousal, sexual receptivity and orgasm.
  • Administration and Dose Ranges
  • Any suitable route of administration may be employed for providing a mammal, especially a human with an effective dosage of a compound of the present invention. For example, oral, rectal, topical, parenteral, ocular, pulmonary, nasal and the like may be employed. Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, aerosols and the like. Preferably compounds of formulas (I) and (II) are administered orally or topically.
  • The effective dosage of active ingredient employed may vary depending on the particular compound employed, the mode of administration, the condition being treated and the severity of the condition being treated. Such dosage may be ascertained readily by a person skilled in the art.
  • When treating cancer cachexia, muscle wasting or anorexia generally satisfactory results are obtained when the compounds of the present invention are administered at a daily dosage of from about 0.001 milligram to about 100 milligrams per kilogram of body weight, preferably given in a single dose or in divided doses two to six times a day, or in sustained release form. In the case of a 70 kg adult human, the total daily dose will generally be from about 0.07 milligrams to about 3500 milligrams. This dosage regimen may be adjusted to provide the optimal therapeutic response.
  • When treating obesity, in conjunction with diabetes and/or hyperglycemia, or alone, generally satisfactory results are obtained when the compounds of the present invention are administered at a daily dosage of from about 0.001 milligram to about 100 milligrams per kilogram of body weight, preferably given in a single dose or in divided doses two to six times a day, or in sustained release form. In the case of a 70 kg adult human, the total daily dose will generally be from about 0.07 milligrams to about 3500 milligrams. This dosage regimen may be adjusted to provide the optimal therapeutic response.
  • When treating diabetes mellitus and/or hyperglycemia, as well as other diseases or disorders for which compounds of formulas I and (II) are useful, generally satisfactory results are obtained when the compounds of the present invention are administered at a daily dosage of from about 0.001 milligram to about 100 milligram per kilogram of animal body weight, preferably given in a single dose or in divided doses two to six times a day, or in sustained release form. In the case of a 70 kg adult human, the total daily dose will generally be from about 0.07 milligrams to about 3500 milligrams. This dosage regimen may be adjusted to provide the optimal therapeutic response.
  • For the treatment of sexual dysfunction, compounds of the present invention are given in a dose range of 0.001 milligram to about 100 milligram per kilogram of body weight, preferably as a single dose orally or as a nasal spray.
  • Formulation
  • The compound of formulas (I) and (II) is preferably formulated into a dosage form prior to administration. Accordingly the present invention also includes a pharmaceutical composition comprising a compound of formulas (I) and (II) and a suitable pharmaceutical carrier.
  • The present pharmaceutical compositions are prepared by known procedures using well-known and readily available ingredients. In making the formulations of the present invention, the active ingredient (a compound of formulas (I) and (II)) is usually mixed with a carrier, or diluted by a carrier, or enclosed within a carrier, which may be in the form of a capsule, sachet, paper or other container. When the carrier serves as a diluent, it may be a solid, semisolid or liquid material which acts as a vehicle, excipient or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosol (as a solid or in a liquid medium), soft and hard gelatin capsules, suppositories, sterile injectable solutions and sterile packaged powders.
  • Some examples of suitable carriers, excipients and diluents include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water syrup, methyl cellulose, methyl and propylhydroxybenzoates, talc, magnesium stearate and mineral oil. The formulations can additionally include lubricating agents, wetting agents, emulsifying and suspending agents, preserving agents, sweetening agents or flavoring agents. The compositions of the invention may be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient.
  • Preparation of Compounds of the Invention
  • When describing the preparation of the present compounds of formulas (I) and (II), the terms “A moiety”, “B moiety” and “C moiety” are used below. This moiety concept is illustrated below:
    Figure US20060241123A1-20061026-C00013
  • Preparation of the compounds of the present invention may be carried out via sequential or convergent synthetic routes. The skilled artisan will recognize that, in general, the three moieties of a compound of formulas (I) and (II) are connected via amide bonds. The skilled artist can, Thereforee, readily envision numerous routes and methods of connecting the three moieties via standard peptide coupling reaction conditions.
  • The phrase “standard peptide coupling reaction conditions” means coupling a carboxylic acid with an amine using an acid activating agent such as EDC, dicyclohexylcarbodiimide and benzotriazol-1-yloxytris(dimethylamino)-phosphonium hexafluorophosphate, in a inert solvent such as DCM, in the presence of a catalyst such as HOBt. The uses of protective groups for amine and carboxylic acids to facilitate the desired reaction and minimize undesired reactions are well documented. Conditions required to remove protecting groups which may be present can be found in Greene, et al., Protective Groups in Organic Synthesis, John Wiley & Sons, Inc., New York, N.Y. 1991.
  • Protecting groups like Z, Boc and Fmoc are used extensively in the synthesis, and their removal conditions are well known to those skilled in the art. For example, removal of Z groups can he achieved by catalytic hydrogenation with hydrogen in the presence of a noble metal or its oxide, such as palladium on activated carbon in a protic solvent, such as ethanol. In cases where catalytic hydrogenation is contraindicated by the presence of other potentially reactive functionality, removal of Z can also be achieved by treatment with a solution of hydrogen bromide in acetic acid, or by treatment with a mixture of TFA and dimethylsulfide. Removal of Boc protecting groups is carried out in a solvent such as methylene chloride, methanol or ethyl acetate with a strong acid, such as TFA or HCl or hydrogen chloride gas.
  • The compounds of formulas (I) and (II), when existing as a diastereomeric mixture, may be separated into diastereomeric pairs of enantiomers by fractional crystallization from a suitable solvent such as methanol, ethyl acetate or a mixture thereof. The pair of enantiomers thus obtained may be separated into individual stereoisomers by conventional means by using an optically active acid as a resolving agent. Alternatively, any enantiomer of a compound of the formulas (I) and (II) may be obtained by stereospecific synthesis using optically pure starting materials or reagents of known configuration.
  • The compounds of formulas (I) and (II) of the present invention can be prepared according to the procedures of the following Schemes and Examples, using appropriate materials and are further exemplified by the following specific examples. Moreover, by utilizing the procedures described herein, in conjunction with ordinary skills in the art, additional compounds of the present invention claimed herein can be readily prepared. The compounds illustrated in the examples are not, however, to be construed as forming the only genus that is considered as the invention. The Examples further illustrate details for the preparation of the compounds of the present invention. Those skilled in the art will readily understand that known variations of the conditions and processes of the following preparative procedures can be used to prepare these compounds. The instant compounds are generally isolated in the form of their pharmaceutically acceptable salts, such as those described previously. The free amine bases corresponding to the isolated salts can be generated by neutralization with a suitable base, such as aqueous sodium hydrogencarbonate, sodium carbonate, sodium hydroxide and potassium hydroxide, and extraction of the liberated amine free base into an organic solvent followed by evaporation. The amine free base isolated in this manner can be further converted into another pharmaceutically acceptable salt by dissolution in an organic solvent followed by addition of the appropriate acid and subsequent evaporation, precipitation or crystallization. All temperatures are degrees Celsius. Mass spectra (MS) were measured by electron-spray ion-mass spectroscopy.
  • In the schemes, preparations and examples below, various reagent symbols and abbreviations have the following meanings:
  • Boc tert-butoxycarbonyl
  • DCM dichloromethane
  • DIPEA diisopropylethylamine
  • DMAP 4-dimethylaminopyridine
  • DMF N,N-dimethylformamide
  • EDC 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • Et ethyl
  • EtOAc ethyl acetate
  • Fmoc 9-fluorenylmethyl-carbamate
  • HOAc acetic acid
  • HOAt 1-hydroxy-7-azabenzotriazole
  • HOBt 1-hydroxybenzotriazole
  • h hour(s)
  • NMM N-methylmorpholine
  • Phe phenylalanine
  • TFA trifluoroacetic acid
  • TEA triethylamine
  • THF tetrahydrofurane
  • Tic 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid
  • TMOF trimethylorthoformate
  • Z benzyloxycarbonyl
    Figure US20060241123A1-20061026-C00014
  • In coupling technique 1, an appropriate “A moiety” (e.g., 4-cyclohexyl-4-[1,2,4]triazol-1-yl-methyl-piperidine) is coupled to “B moiety” (e.g., L-Boc-p-Cl-Phe-OH) in the presence of EDC/HOBt followed by Boc deprotection. The coupled AB compound is then coupled to an appropriate “C moiety” followed by deprotection of Boc group and salt formation. Alternatively, when “C moiety” is not protected with Boc group, the final compound can be obtained without the deprotection step.
  • In coupling technique 2, an appropriate “AB moiety” is coupled to an appropriate “C moiety” in the presence of EDC/HOBt followed by deprotection of Boc group and salt formation. Alternatively, when “C moiety” is not protected with Boc group, the final compound can be obtained without the deprotection step.
  • In coupling technique 3, an appropriate “BC moiety” is coupled to an appropriate “A moiety” in the presence of EDC/HOBt followed by deprotection of Boc group and salt formation. Alternatively, when “C moiety” is not protected with Boc group, the final compound can be obtained without the deprotection step.
  • For coupling of A with Boc-B—OH, EDC/HOAt, EDC/HOBt or DCC/HOBt can be used.
  • Generally, the starting material of Boc-protected piperazine or piperidine (A moiety) can be deprotected in the presence of TFA/CH2Cl2, HCl/EtOAc, HCl/dioxane or HCl in MeOH/Et2O with or without a cation scavenger, such as dimethyl sulfide (DMS) before being subjected to the coupling procedure. It can be free-based before being subjected to the coupling procedure or in some cases used as the salt.
  • A suitable solvent, such as CH2Cl2; DMF, THF or a mixture of the above solvents, can be used for the coupling procedure. A suitable base includes triethylamine (TEA), diisopropyethylamine (DIPEA), N-methymorpholine (NMM), collidine or 2,6-lutidine.
  • A base may not be needed when EDC/HOBt is used.
  • Generally after the reaction is completed, the reaction mixture can be diluted with an appropriate organic solvent, such as EtOAc, CH2Cl2 or Et2O, which is then washed with aqueous solutions, such as water, HCl, NaHSO4, bicarbonate, NaH2PO4, phosphate buffer (pH 7), brine or any combination thereof. The reaction mixture can be concentrated and then be partitioned between an appropriate organic solvent and an aqueous solution. The reaction mixture can be concentrated and subjected to chromatography without aqueous workup.
  • Protecting groups such as Boc, Z, Fmoc and CF3CO can be deprotected in the presence of H2/Pd—C, TFA/DCM, HCl/EtOAc, HCl/dioxane, HCl in MeOH/Et2O, NH3/MeOH or TBAF, with or without a cation scavenger, such as thioanisole, ethane thiol and dimethyl sulfide (DMS). The deprotected amines can be used as the resulting salt or are free-based by dissolving in DCM and washing with aqueous bicarbonate or aqueous NaOH. The deprotected amines can also be free-based by ion exchange chromatography.
  • The “A moieties” can be prepared as described in the corresponding literature: 4-Cyclohexyl-4-[1,2,4]triazol-1-ylmethyl-piperidine (WO0074679), 4-cyclohexyl-piperidine-4-carboxylic acid tert-butylamide (WO0170708), 1,2-dihydro-1-methanesulfonylspiro-[3H-indole-3,4′-piperidine] (US005536716), 3a-benzyl-2-methyl-2,3a,4,5,6,7-hexahydro-pyrazolo[4,3-c]pyridin-3-one (WO9858949) and N-(2-piperazin-1-yl-benzyl)-methane-sulfonamide, 1-(2-[1,2,4]triazol-1-ylmethyl-phenyl)-piperazine, N-(2-piperazin-1-yl-phenyl)-isobutyramide, N-(2-piperazin-1-yl-phenyl)-methanesulfonamide, (2-piperazin-1-yl-phenyl)-piperidin-1-yl-methanone, N,N-diethyl-2-piperazin-1-yl-benzamide (WO02059108).
  • The following “A moieties” were prepared in an analogues way:
  • N-(2-piperazin-1-yl-phenyl)-benzamide, (4-methyl-piperazin-1-yl)-(2-piperazin-1-yl-phenyl)-methanone, morpholin-4-yl-(2-piperazin-1-yl-phenyl)-methanone, (2-piperazin-1-yl-phenyl)-pyrrolidin-1-yl-methanone, N-(2-methoxy-ethyl)-2-piperazin-1-yl-benzamide, N-(2-dimethylamino-ethyl)-2-piperazin-1-yl-benzamide, 2-piperazin-1-yl-N-(2,2,2-trifluoro-ethyl)-benzamide, N-cyclopropyl-2-piperazin-1-yl-benzamide, 4-methyl-N-(2-piperazin-1-yl-phenyl)-benzene-sulfonamide, 4-bromo-2-piperazin-1-yl-benzonitrile (WO02059108) and 4 cyclohexyl-piperidine-4-carboxylic acid methyl ester (WO0170708).
    Reaction Schemes for Preparation of “C Moiety”
    Figure US20060241123A1-20061026-C00015
  • As shown in Reaction Scheme 2, ethyl 3-bromo-4-oxochromene-2-carboxylate 1 (J. Chem. Soc. Perkin Trans. I 1986, 1643-1649) can be reacted with amines, with or without a base such as K2CO3, in an appropriate solvent such as MeCN, to form products 2 which are subsequently treated with a reagent such as HBr/HOAc, to form carboxylic acids 3. When R8 is hydrogen, the free amine can be protected with a reagent such as Boc2O in the presence of TEA and DMAP in an appropriate solvent.
    Figure US20060241123A1-20061026-C00016
  • As shown in Reaction Scheme 3, ethyl 4-oxo-1,4-dihydro-quinoline-2-carboxylates 5 (Bioorg. Med. Chem. Lett. 2000, 10, 1487-1490) can be converted into the corresponding acids 6 by an appropriate reactant such as HBr/HOAc.
    Figure US20060241123A1-20061026-C00017
  • As shown in Reaction Scheme 4, substituted phenols 7 can be reacted with triethylamine followed by dimethyl acetylendicarboxylate in diethyl ether to yield compounds 8 (Aust. J. Chem. 1995, 48, 677-686). Saponification of the latter with aqueous sodium hydroxide leads to acids 9 which are subsequently cyclized to the chromone-2-carboxylic acids 10 using concentrated sulfuric acid in acetyl chloride.
    Figure US20060241123A1-20061026-C00018
  • As shown in Reaction Scheme 5,2′-hydroxyacetophenones 11 can be reacted with diethyl oxalate 12 in the presence of a base such as sodium methoxide in an appropriate solvent such as methanol or benzene followed by treatment with an acid such as hydrochloric acid to yield chromone-2-carboxylic acid esters 13 (J. Indian Chem. Soc. 1986, 63, 600-602). The esters can be cleaved using basic conditions such as sodium bicarbonate in water or acidic conditions such as polyphosphoric acid at an appropriate temperature to the corresponding acids 10.
    Figure US20060241123A1-20061026-C00019
  • As shown in Reaction Scheme 6, methoxy-substituted chromone-2-carboxylic acids can be demethylates with reagents such as hydroiodic acid in an appropriate solvent such as glacial acetic acid to yield the corresponding hydroxy-substituted chromone-2-carboxylic acids.
  • 5,7-Dihydroxychromane-2-carboxylic acid was prepared as described in the literature (OPPI Briefs 1991, 23, 390-392).
  • The following describes the detailed examples of the invention
    Figure US20060241123A1-20061026-C00020
  • Synthesis Scheme for Example 1
  • Figure US20060241123A1-20061026-C00021
  • Synthesis Scheme for Example 211
  • The following examples are provided to illustrate the invention and are not limiting the scope of the invention in any manner.
  • EXAMPLE 1
  • Figure US20060241123A1-20061026-C00022
  • To chromone-2-carboxylic acid (17 mg) in DCM (2 ml) was added intermediate 1b) (39 mg), N-methylmorpholine (14 μl), HOBt (14 mg) and stirred for 20 min. EDC (23 mg) was added and stirring was continued for 1 h. An additional amount of N-methylmorpholine (8 μl) was added and stirred overnight. The reaction mixture was poured into water (5 ml) and the organic phase was separated. The aqueous phase was extracted two times with DCM. The combined organic phases were washed with 0.5 N HCl and saturated sodium bicarbonate solution, dried over Na2SO4 and concentrated to yield the product which was purified by column chromatography.
  • colorless solid
  • Rf=0.52 (EtOAc); Mp. 137-147° C.
  • The required intermediates can be synthesized in the following way:
    Intermediate 1a):
    Figure US20060241123A1-20061026-C00023
  • To Boc-D-4-chlorophenylalanine (82 mg) in DCM (5 ml) was added the amine hydrochloride (preparation in US005536716) (62 mg), N-methylmorpholine (42 μl), HOBt (48 mg) and stirred for 20 min. EDC (72 mg) was added and stirring was continued for 1 h. An additional amount of N-methylmorpholine (20 μl) was added and stirred overnight. The reaction mixture was poured into water (5 ml) and the organic phase was separated. The aqueous phase was extracted two times with DCM. The combined organic phases were washed with 0.5 N HCl and saturated sodium bicarbonate solution, dried over Na2SO4 and concentrated in vacuo. Purification by column chromatography yielded the title compound.
    Intermediate 1b):
    Figure US20060241123A1-20061026-C00024
  • To the Boc-protected amine from 1a) (86 mg) in DCM (5 ml) was added TFA (1 ml) and stirred at room temperature for 90 min. Additional TFA (1 ml) was added and stirred for 10 min. The reaction mixture was diluted with DCM (10 ml) and carefully basified by pouring into 10% aqueous sodium carbonate solution (20 ml). The organic layer was separated and the aqueous layer was further extracted three times with DCM. The combined organics were washed with water and brine, dried over Na2SO4, concentrated to give a white solid.
  • For prolonged storage, the free base was converted into the corresponding hydrochloride. The free base was dissolved in DCM (5 ml) and app. 1 M HCl in ether (10 ml) was added. The precipitate was filtered and the residue was washed three times with ether and dried under reduced pressure to yield the desired compound.
  • The following examples can be prepared in a similar way:
  • EXAMPLE 2
  • Figure US20060241123A1-20061026-C00025

    colorless solid
    Rf=0.52 (EtOAc); Mp. 139-160° C.
  • EXAMPLE 3
  • Figure US20060241123A1-20061026-C00026

    white solid
    Rf=0.56 (EtOAc); Mp. 203-204° C.
  • EXAMPLE 4
  • Figure US20060241123A1-20061026-C00027

    white solid
    Rf=0.75 (EtOAc/ethanol 3:1); Mp. 166-168° C.
  • EXAMPLE 5
  • Figure US20060241123A1-20061026-C00028

    white solid
    Rf=0.61 (EtOAc); Mp. 94° C.
  • EXAMPLE 6
  • Figure US20060241123A1-20061026-C00029

    white solid
    Rf=0.71 (EtOAc); Mp. 147-156° C.
  • EXAMPLE 7
  • Figure US20060241123A1-20061026-C00030

    pale yellow solid
    Rf=0.61 (EtOAc); Mp. 119-126° C.
  • EXAMPLE 8
  • Figure US20060241123A1-20061026-C00031

    white solid
    Rf=0.62 (EtOAc); Mp. 136-140° C.
  • EXAMPLE 9
  • Figure US20060241123A1-20061026-C00032

    white solid
    Rf=0.77 (EtOAc); Mp. 157-161° C.
  • EXAMPLE 10
  • Figure US20060241123A1-20061026-C00033

    white solid
    Rf=0.77 (EtOAc); Mp. 160-167° C.
  • EXAMPLE 11
  • Figure US20060241123A1-20061026-C00034

    white solid
    Rf=0.74 (EtOAc); Mp. 140-149° C.
  • EXAMPLE 12
  • Figure US20060241123A1-20061026-C00035

    white solid
    Rf=0.74 (EtOAc); Mp. 128-135° C.
  • EXAMPLE 13
  • Figure US20060241123A1-20061026-C00036

    white solid
    Rf=0.74 (EtOAc); Mp. 107-118° C.
  • EXAMPLE 14
  • Figure US20060241123A1-20061026-C00037

    colorless glassy solid
    Rf=0.67 (EtOAc/ethanol 3:1); Mp. 126-135° C.
  • EXAMPLE 15
  • Figure US20060241123A1-20061026-C00038

    white solid
    Rf=0.67 (EtOAc/ethanol 3:1); Mp. 117-126° C.
  • EXAMPLE 16
  • Figure US20060241123A1-20061026-C00039

    white solid
    Rf=0.77 (EtOAc/ethanol 3:1); Mp. 147-157° C.
  • EXAMPLE 17
  • Figure US20060241123A1-20061026-C00040

    white solid
    Rf=0.65 (EtOAc/ethanol 3:1); Mp. 136-141° C.
  • EXAMPLE 18
  • Figure US20060241123A1-20061026-C00041

    white solid
    Rf=0.70 (EtOAc/ethanol 3:1); Mp. 124-132° C.
  • EXAMPLE 19
  • Figure US20060241123A1-20061026-C00042

    white solid
    Rf=0.75 (EtOAc/ethanol 3:1); Mp. 147-150° C.
  • EXAMPLE 20
  • Figure US20060241123A1-20061026-C00043

    off-white solid
    Rf=0.58 (EtOAc/ethanol 3:1); Mp. 120-125° C.
  • EXAMPLE 21
  • Figure US20060241123A1-20061026-C00044

    white solid
    Rf=0.56 (EtOAc/ethanol 3:1); Mp. 139-144° C.
  • EXAMPLE 22
  • Figure US20060241123A1-20061026-C00045

    white solid
    Rf=0.54 (EtOAc/ethanol 3:1); Mp. 132-140° C.
  • EXAMPLE 23
  • Figure US20060241123A1-20061026-C00046

    off-white solid
    Rf=0.56 (EtOAc/ethanol 3:1); Mp. 154-162° C.
  • EXAMPLE 24
  • Figure US20060241123A1-20061026-C00047

    off-white solid
    Rf=0.69 (EtOAc/ethanol 3:1); Mp. 117-130° C.
  • EXAMPLE 25
  • Figure US20060241123A1-20061026-C00048

    white solid
    Rf=0.64 (EtOAc/ethanol 3:1); Mp. 137-142° C.
  • EXAMPLE 26
  • Figure US20060241123A1-20061026-C00049

    white solid
    Rf=0.53 (EtOAc/ethanol 3:1); Mp. 144-157° C.
  • EXAMPLE 27
  • Figure US20060241123A1-20061026-C00050

    beige solid
    Rf=0.66 (EtOAc/ethanol 3:1); Mp. 166-173° C.
  • EXAMPLE 28
  • Figure US20060241123A1-20061026-C00051

    white solid
    Rf=0.60 (EtOAc/ethanol 3:1); Mp. 142-146° C.
  • EXAMPLE 29
  • Figure US20060241123A1-20061026-C00052

    white solid
    Rf=0.65 (EtOAc/ethanol 3:1); Mp. 130-141° C.
  • EXAMPLE 30
  • Figure US20060241123A1-20061026-C00053

    white solid
    Rf=0.76 (EtOAc/ethanol 3:1); Mp. 116-124° C.
  • EXAMPLE 31
  • Figure US20060241123A1-20061026-C00054

    white solid
    Rf=0.69 (EtOAc/ethanol 3:1); Mp. 119-126° C.
  • EXAMPLE 32
  • Figure US20060241123A1-20061026-C00055
  • EXAMPLE 33
  • Figure US20060241123A1-20061026-C00056
  • EXAMPLE 34
  • Figure US20060241123A1-20061026-C00057
  • EXAMPLE 35
  • Figure US20060241123A1-20061026-C00058
  • EXAMPLE 36
  • Figure US20060241123A1-20061026-C00059

    white solid
    Rf=0.83 (EtOAc/ethanol 3:1); Mp. 122-130° C.
  • EXAMPLE 37
  • Figure US20060241123A1-20061026-C00060

    white solid
    Rf=0.77 (EtOAc/ethanol 3:1); Mp. 123-134° C.
  • EXAMPLE 38
  • Figure US20060241123A1-20061026-C00061

    white solid
    Rf=0.78 (EtOAc/ethanol 3:1); Mp. 123-132° C.
  • EXAMPLE 39
  • Figure US20060241123A1-20061026-C00062
  • EXAMPLE 40
  • Figure US20060241123A1-20061026-C00063
  • EXAMPLE 41
  • Figure US20060241123A1-20061026-C00064
  • EXAMPLE 42
  • Figure US20060241123A1-20061026-C00065
  • EXAMPLE 43
  • Figure US20060241123A1-20061026-C00066
  • EXAMPLE 44
  • Figure US20060241123A1-20061026-C00067
  • EXAMPLE 45
  • Figure US20060241123A1-20061026-C00068
  • EXAMPLE 46
  • Figure US20060241123A1-20061026-C00069
  • EXAMPLE 47
  • Figure US20060241123A1-20061026-C00070
  • EXAMPLE 48
  • Figure US20060241123A1-20061026-C00071
  • EXAMPLE 49
  • Figure US20060241123A1-20061026-C00072
  • EXAMPLE 50
  • Figure US20060241123A1-20061026-C00073
  • EXAMPLE 51
  • Figure US20060241123A1-20061026-C00074
  • EXAMPLE 52
  • Figure US20060241123A1-20061026-C00075
  • EXAMPLE 53
  • Figure US20060241123A1-20061026-C00076
  • EXAMPLE 54
  • Figure US20060241123A1-20061026-C00077
  • EXAMPLE 55
  • Figure US20060241123A1-20061026-C00078
  • EXAMPLE 56
  • Figure US20060241123A1-20061026-C00079
  • EXAMPLE 57
  • Figure US20060241123A1-20061026-C00080
  • EXAMPLE 58
  • Figure US20060241123A1-20061026-C00081

    white solid
    Rf=0.62 (EtOAc/ethanol 3:1); Mp. 154-162° C.
  • EXAMPLE 59
  • Figure US20060241123A1-20061026-C00082

    white solid
    Rf=0.66 (EtOAc/ethanol 3:1); Mp. 143-150° C.
  • EXAMPLE 60
  • Figure US20060241123A1-20061026-C00083

    white solid
    Rf=0.68 (EtOAc/ethanol 3:1); Mp. 118-124° C.
  • EXAMPLE 61
  • Figure US20060241123A1-20061026-C00084

    white solid
    Rf=0.66 (EtOAc/ethanol 3:1); Mp. 132-142° C.
  • EXAMPLE 62
  • Figure US20060241123A1-20061026-C00085

    white solid
    Rf=0.66 (EtOAc/ethanol 3:1); Mp. 132-142° C.
  • EXAMPLE 63
  • Figure US20060241123A1-20061026-C00086

    white solid
    Rf=0.54 (EtOAc); Mp. 140-163° C.
  • EXAMPLE 64
  • Figure US20060241123A1-20061026-C00087

    white solid
    Rf=0.54 (EtOAc); Mp. 149-158° C.
  • EXAMPLE 65
  • Figure US20060241123A1-20061026-C00088

    white solid
    Rf=0.58 (EtOAc); Mp. 136-148° C.
  • EXAMPLE 66
  • Figure US20060241123A1-20061026-C00089

    white solid
    Rf=0.58 (EtOAc); Mp. 139-151° C.
  • EXAMPLE 67
  • Figure US20060241123A1-20061026-C00090

    white solid
    Rf=0.83 (EtOAc); Mp. 139-146° C.
  • EXAMPLE 68
  • Figure US20060241123A1-20061026-C00091

    white solid
    Rf=0.71 (EtOAc); Mp. 170-177° C.
  • EXAMPLE 69
  • Figure US20060241123A1-20061026-C00092

    white solid
    Rf=0.82 (EtOAc); Mp. 157-166° C.
  • EXAMPLE 70
  • Figure US20060241123A1-20061026-C00093

    white solid
    Rf=0.74 (EtOAc); Mp. 127-131° C.
  • EXAMPLE 71
  • Figure US20060241123A1-20061026-C00094

    white solid
    Rf=0.72 (EtOAc); Mp. 141-148° C.
  • EXAMPLE 72
  • Figure US20060241123A1-20061026-C00095

    white solid
    Rf=0.63 (EtOAc); Mp. 137-140° C.
  • EXAMPLE 73
  • Figure US20060241123A1-20061026-C00096
  • EXAMPLE 74
  • Figure US20060241123A1-20061026-C00097
  • EXAMPLE 75
  • Figure US20060241123A1-20061026-C00098
  • EXAMPLE 76
  • Figure US20060241123A1-20061026-C00099
  • EXAMPLE 77
  • Figure US20060241123A1-20061026-C00100
  • EXAMPLE 78
  • Figure US20060241123A1-20061026-C00101
  • EXAMPLE 79
  • Figure US20060241123A1-20061026-C00102
  • EXAMPLE 80
  • Figure US20060241123A1-20061026-C00103
  • EXAMPLE 81
  • Figure US20060241123A1-20061026-C00104
  • EXAMPLE 82
  • Figure US20060241123A1-20061026-C00105
  • EXAMPLE 83
  • Figure US20060241123A1-20061026-C00106
  • EXAMPLE 84
  • Figure US20060241123A1-20061026-C00107
  • EXAMPLE 85
  • Figure US20060241123A1-20061026-C00108
  • EXAMPLE 86
  • Figure US20060241123A1-20061026-C00109
  • EXAMPLE 87
  • Figure US20060241123A1-20061026-C00110
  • EXAMPLE 88
  • Figure US20060241123A1-20061026-C00111
  • EXAMPLE 89
  • Figure US20060241123A1-20061026-C00112
  • EXAMPLE 90
  • Figure US20060241123A1-20061026-C00113
  • EXAMPLE 91
  • Figure US20060241123A1-20061026-C00114
  • EXAMPLE 92
  • Figure US20060241123A1-20061026-C00115
  • EXAMPLE 93
  • Figure US20060241123A1-20061026-C00116
  • EXAMPLE 94
  • Figure US20060241123A1-20061026-C00117
  • EXAMPLE 95
  • Figure US20060241123A1-20061026-C00118
  • EXAMPLE 96
  • Figure US20060241123A1-20061026-C00119
  • EXAMPLE 97
  • Figure US20060241123A1-20061026-C00120
  • EXAMPLE 98
  • Figure US20060241123A1-20061026-C00121
  • EXAMPLE 99
  • Figure US20060241123A1-20061026-C00122
  • EXAMPLE 100
  • Figure US20060241123A1-20061026-C00123
  • EXAMPLE 101
  • Figure US20060241123A1-20061026-C00124
  • EXAMPLE 102
  • Figure US20060241123A1-20061026-C00125
  • EXAMPLE 103
  • Figure US20060241123A1-20061026-C00126
  • EXAMPLE 104
  • Figure US20060241123A1-20061026-C00127
  • EXAMPLE 105
  • Figure US20060241123A1-20061026-C00128
  • EXAMPLE 106
  • Figure US20060241123A1-20061026-C00129
  • EXAMPLE 107
  • Figure US20060241123A1-20061026-C00130
  • EXAMPLE 108
  • Figure US20060241123A1-20061026-C00131
  • EXAMPLE 109
  • Figure US20060241123A1-20061026-C00132

    Yellow Solid
    Rf=0.79 (EtOAc).
  • EXAMPLE 110
  • Figure US20060241123A1-20061026-C00133

    white solid
    Rf=0.63 (EtOAc); Mp. 160-174° C.
  • EXAMPLE 111
  • Figure US20060241123A1-20061026-C00134

    white solid
    Rf=0.72 (EtOAc/ethanol 3:1); Mp. 113-131° C.
  • EXAMPLE 112
  • Figure US20060241123A1-20061026-C00135

    white solid
    Rf=0.85 (EtOAc); Mp. 131-139° C.
  • EXAMPLE 113
  • Figure US20060241123A1-20061026-C00136

    white solid
    Rf=0.77 (EtOAc/ethanol 3:1); Mp. 135-141° C.
  • EXAMPLE 114
  • Figure US20060241123A1-20061026-C00137

    white solid
    Rf=0.32 (EtOAc); Mp. 141-147° C.
  • EXAMPLE 115
  • Figure US20060241123A1-20061026-C00138

    white solid
    Rf=0.30 (EtOAc); Mp. 129-145° C.
  • EXAMPLE 116
  • Figure US20060241123A1-20061026-C00139

    white solid
    Rf=0.39 (EtOAc); Mp. 210-215° C.
  • EXAMPLE 117
  • Figure US20060241123A1-20061026-C00140

    white solid
    Rf=0.44 (EtOAc/ethanol 3:1); Mp. 148-154° C.
  • EXAMPLE 118
  • Figure US20060241123A1-20061026-C00141
  • EXAMPLE 119
  • Figure US20060241123A1-20061026-C00142
  • EXAMPLE 120
  • Figure US20060241123A1-20061026-C00143

    white solid
    Rf=0.31 (EtOAc); Mp. 141-145° C.
  • EXAMPLE 121
  • Figure US20060241123A1-20061026-C00144

    white solid
    Rf=0.22 (EtOAc); Mp. 150-153° C.
  • EXAMPLE 122
  • Figure US20060241123A1-20061026-C00145

    white solid
    Rf=0.36 (EtOAc); Mp. 134-152° C.
  • EXAMPLE 123
  • Figure US20060241123A1-20061026-C00146

    white solid
    Rf=0.11 (EtOAc); Mp. 140-152° C.
  • EXAMPLE 124
  • Figure US20060241123A1-20061026-C00147

    beige solid
    Rf=0.30 (EtOAc); Mp. 163-171° C.
  • EXAMPLE 125
  • Figure US20060241123A1-20061026-C00148
  • EXAMPLE 126
  • Figure US20060241123A1-20061026-C00149

    white solid
    Rf=0.44 (EtOAc); Mp. 139-144° C.
  • EXAMPLE 127
  • Figure US20060241123A1-20061026-C00150
  • EXAMPLE 128
  • Figure US20060241123A1-20061026-C00151
  • EXAMPLE 129
  • Figure US20060241123A1-20061026-C00152
  • EXAMPLE 130
  • Figure US20060241123A1-20061026-C00153
  • EXAMPLE 131
  • Figure US20060241123A1-20061026-C00154
  • EXAMPLE 131
  • Figure US20060241123A1-20061026-C00155

    white solid
    Rf=0.47 (EtOAc); Mp. 130-135° C.
  • EXAMPLE 133
  • Figure US20060241123A1-20061026-C00156

    white solid
    Rf=0.18 (EtOAc); Mp. 143-150° C.
  • EXAMPLE 134
  • Figure US20060241123A1-20061026-C00157
  • EXAMPLE 135
  • Figure US20060241123A1-20061026-C00158
  • EXAMPLE 136
  • Figure US20060241123A1-20061026-C00159
  • EXAMPLE 137
  • Figure US20060241123A1-20061026-C00160
  • EXAMPLE 138
  • Figure US20060241123A1-20061026-C00161
  • EXAMPLE 139
  • Figure US20060241123A1-20061026-C00162
  • EXAMPLE 140
  • Figure US20060241123A1-20061026-C00163
  • EXAMPLE 141
  • Figure US20060241123A1-20061026-C00164
  • EXAMPLE 142
  • Figure US20060241123A1-20061026-C00165
  • EXAMPLE 143
  • Figure US20060241123A1-20061026-C00166
  • EXAMPLE 144
  • Figure US20060241123A1-20061026-C00167
  • EXAMPLE 145
  • Figure US20060241123A1-20061026-C00168
  • EXAMPLE 146
  • Figure US20060241123A1-20061026-C00169
  • EXAMPLE 147
  • Figure US20060241123A1-20061026-C00170
  • EXAMPLE 148
  • Figure US20060241123A1-20061026-C00171
  • EXAMPLE 149
  • Figure US20060241123A1-20061026-C00172
  • EXAMPLE 150
  • Figure US20060241123A1-20061026-C00173
  • EXAMPLE 151
  • Figure US20060241123A1-20061026-C00174
  • EXAMPLE 152
  • Figure US20060241123A1-20061026-C00175
  • EXAMPLE 153
  • Figure US20060241123A1-20061026-C00176
  • EXAMPLE 154
  • Figure US20060241123A1-20061026-C00177
  • EXAMPLE 155
  • Figure US20060241123A1-20061026-C00178
  • EXAMPLE 156
  • Figure US20060241123A1-20061026-C00179

    white solid
    Rf=0.51 (EtOAc); Mp. 125-136° C.
  • EXAMPLE 157
  • Figure US20060241123A1-20061026-C00180

    white solid
    Rf=0.45 (EtOAc); Mp. 170-188° C.
  • EXAMPLE 158
  • Figure US20060241123A1-20061026-C00181

    white solid
    Rf=0.06 (EtOAc/ethanol 3:1); Mp. 160-165° C.
  • EXAMPLE 159
  • Figure US20060241123A1-20061026-C00182
  • EXAMPLE 160
  • Figure US20060241123A1-20061026-C00183
  • EXAMPLE 161
  • Figure US20060241123A1-20061026-C00184
  • EXAMPLE 162
  • Figure US20060241123A1-20061026-C00185
  • EXAMPLE 163
  • Figure US20060241123A1-20061026-C00186
  • EXAMPLE 164
  • Figure US20060241123A1-20061026-C00187
  • EXAMPLE 165
  • Figure US20060241123A1-20061026-C00188
  • EXAMPLE 166
  • Figure US20060241123A1-20061026-C00189
  • EXAMPLE 167
  • Figure US20060241123A1-20061026-C00190
  • EXAMPLE 168
  • Figure US20060241123A1-20061026-C00191
  • EXAMPLE 169
  • Figure US20060241123A1-20061026-C00192
  • EXAMPLE 170
  • Figure US20060241123A1-20061026-C00193
  • EXAMPLE 171
  • Figure US20060241123A1-20061026-C00194
  • EXAMPLE 172
  • Figure US20060241123A1-20061026-C00195
  • EXAMPLE 173
  • Figure US20060241123A1-20061026-C00196
  • EXAMPLE 174
  • Figure US20060241123A1-20061026-C00197
  • EXAMPLE 175
  • Figure US20060241123A1-20061026-C00198
  • EXAMPLE 176
  • Figure US20060241123A1-20061026-C00199
  • EXAMPLE 177
  • Figure US20060241123A1-20061026-C00200
  • EXAMPLE 178
  • Figure US20060241123A1-20061026-C00201
  • EXAMPLE 179
  • Figure US20060241123A1-20061026-C00202
  • EXAMPLE 180
  • Figure US20060241123A1-20061026-C00203
  • EXAMPLE 181
  • Figure US20060241123A1-20061026-C00204
  • EXAMPLE 182
  • Figure US20060241123A1-20061026-C00205
  • EXAMPLE 183
  • Figure US20060241123A1-20061026-C00206
  • EXAMPLE 184
  • Figure US20060241123A1-20061026-C00207
  • EXAMPLE 185
  • Figure US20060241123A1-20061026-C00208
  • EXAMPLE 186
  • Figure US20060241123A1-20061026-C00209
  • EXAMPLE 187
  • Figure US20060241123A1-20061026-C00210
  • EXAMPLE 188
  • Figure US20060241123A1-20061026-C00211
  • EXAMPLE 189
  • Figure US20060241123A1-20061026-C00212
  • EXAMPLE 190
  • Figure US20060241123A1-20061026-C00213
  • EXAMPLE 191
  • Figure US20060241123A1-20061026-C00214
  • EXAMPLE 192
  • Figure US20060241123A1-20061026-C00215
  • EXAMPLE 193
  • Figure US20060241123A1-20061026-C00216
  • EXAMPLE 194
  • Figure US20060241123A1-20061026-C00217
  • EXAMPLE 195
  • Figure US20060241123A1-20061026-C00218
  • EXAMPLE 196
  • Figure US20060241123A1-20061026-C00219
  • EXAMPLE 197
  • Figure US20060241123A1-20061026-C00220
  • EXAMPLE 198
  • Figure US20060241123A1-20061026-C00221
  • EXAMPLE 199
  • Figure US20060241123A1-20061026-C00222
  • EXAMPLE 200
  • Figure US20060241123A1-20061026-C00223
  • EXAMPLE 201
  • Figure US20060241123A1-20061026-C00224

    white solid
    Rf=0.45 (EtOAc); Mp. 196-206° C.
  • EXAMPLE 202
  • Figure US20060241123A1-20061026-C00225

    colorless glassy solid
    Rf=0.69 (EtOAc/ethanol 3:1); Mp. 100-103° C.
  • EXAMPLE 203
  • Figure US20060241123A1-20061026-C00226

    colorless glassy solid
    Rf=0.06 (EtOAc/ethanol 3:1); Mp. 96-99° C.
  • EXAMPLE 204
  • Figure US20060241123A1-20061026-C00227

    white solid
    Rf=0.66 (EtOAc/ethanol 3:1); Mp. 138-147° C.
  • EXAMPLE 205
  • Figure US20060241123A1-20061026-C00228

    white solid
    Rf=0.86 (EtOAc/ethanol 3:1); Mp. 119-124° C.
  • EXAMPLE 206
  • Figure US20060241123A1-20061026-C00229

    white solid
    Rf=0.28 (EtOAc/hexane 1:1); Mp. 152-155° C.
  • EXAMPLE 207
  • Figure US20060241123A1-20061026-C00230

    white solid
    Rf=0.68 (EtOAc/ethanol 3:1); Mp. 144-152° C.
  • EXAMPLE 208
  • Figure US20060241123A1-20061026-C00231

    white solid
    Rf=0.45 (EtOAc); Mp. 134-146° C.
  • EXAMPLE 209
  • Figure US20060241123A1-20061026-C00232

    white solid
    Rf=0.45 (EtOAc); Mp. 137-143° C.
  • EXAMPLE 210
  • Figure US20060241123A1-20061026-C00233

    pale yellow solid
    Rf=0.70 (EtOAc/ethanol 3:1); Mp. 144-153° C.
  • EXAMPLE 211
  • Figure US20060241123A1-20061026-C00234
  • To Boc-protected intermediate 211c) (32 mg) was added hydrogen chloride, 4.0 M sol. in 1,4-dioxane (10 ml) and the solution was stirred for 90 min at room temperature. The solvent was removed under reduced pressure. The residue was dissolved in DCM and treated with diethyl ether. The precipitate was filtered off to yield the title compound as a solid.
  • white solid
  • Mp. 160-175° C.
  • The required intermediates can be synthesized in the following way:
    Intermediate 211a):
    Figure US20060241123A1-20061026-C00235
  • To Boc-(S)-3-amino-(4-chloro-phenyl)-butyric acid (170 mg) in DCM (6 ml) was added 1,2-dihydro-1-methanesulfonylspiro-[3H-indole-3,4′-piperidine] hydro-chloride (179 mg), N-methylmorpholine (83 μl), HOBt (140 mg) and stirred for 25 min. EDC (144 mg) was added and stirring was continued for 1 h. An additional amount of N-methylmorpholine (30 μl) was added and stirred overnight. The reaction mixture was poured into water (5 ml) and the organic phase was separated. The aqueous phase was extracted two times with DCM. The combined organic phases were washed with 0.5 N HCl and saturated sodium bicarbonate solution, dried over Na2SO4 and concentrated. The crude product was slurried in ethanol/ethyl acetate and filtrated to yield the desired compound after filtration.
  • white solid
  • Rf=0.47 (EtOAc).
  • Intermediate 211b):
    Figure US20060241123A1-20061026-C00236
  • To intermediate 211a) (210 mg) in DCM (5 ml) was added TFA (1 ml) and stirred at room temperature for 2.5 h. Additional TFA (1 ml) was added and stirred for 1 h. The reaction mixture was diluted with DCM (5 ml) and carefully basified by pouring it into 10% aqueous sodium carbonate solution (10 ml). The organic layer was separated and the aqueous layer was further extracted twice with DCM. The combined organics were washed with water and brine, dried over Na2SO4 and concentrated to give colorless glassy solid.
  • The crude product was dissolved in DCM (5 ml) and app. 1 M HCl in ether (10 ml) was added. The precipitate was filtered and the residue was washed three times with ether and dried under reduced pressure to yield the title compound.
  • white solid
  • Mp. 160-175° C.
  • Intermediate 211c):
    Figure US20060241123A1-20061026-C00237
  • To intermediate 211 b) (23 mg) and DCM (1 ml) was added (R)-Boc-1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid (16 mg), HOBt (9 mg) and EDC (15 mg) and stirred overnight. The reaction mixture was poured into water (5 ml) and the organic layer was separated. The aqueous layer was extracted twice with DCM. The combined organics were washed with 0.5 N HCl (5 ml) and saturated sodium bicarbonate solution, dried over Na2SO4 and concentrated. The crude product was purified by column chromatography.
  • colorless solid
  • Rf=0.76 (EtOAc/ethanol 3:1).
  • The following examples can be prepared in a similar way:
  • EXAMPLE 212
  • Figure US20060241123A1-20061026-C00238

    white solid
    Mp. 162-177° C.
  • EXAMPLE 213
  • Figure US20060241123A1-20061026-C00239

    white solid
    Mp. 192-200° C.
  • EXAMPLE 214
  • Figure US20060241123A1-20061026-C00240

    white solid
    Mp. 189-201° C.
  • EXAMPLE 215
  • Figure US20060241123A1-20061026-C00241

    white solid
    Mp. 161-170° C.
  • EXAMPLE 216
  • Figure US20060241123A1-20061026-C00242

    white solid
    Mp. 180-185° C.
  • EXAMPLE 217
  • Figure US20060241123A1-20061026-C00243

    white solid
    Mp. 190-199° C.
  • EXAMPLE 218
  • Figure US20060241123A1-20061026-C00244

    white solid
    Mp. 198-206° C.
  • EXAMPLE 219
  • Figure US20060241123A1-20061026-C00245

    white solid
    Mp. 207-238° C.
  • EXAMPLE 220
  • Figure US20060241123A1-20061026-C00246

    white solid
    Mp. 225-253° C.
  • EXAMPLE 221
  • Figure US20060241123A1-20061026-C00247

    white solid
    Mp. 176-187° C.
  • EXAMPLE 222
  • Figure US20060241123A1-20061026-C00248

    white solid
    Mp. 148° C.
  • EXAMPLE 223
  • Figure US20060241123A1-20061026-C00249

    white solid
    Mp. 150-155° C.
  • EXAMPLE 224
  • Figure US20060241123A1-20061026-C00250

    white solid
    Mp. 150-156° C.
  • EXAMPLE 225
  • Figure US20060241123A1-20061026-C00251

    white solid
    Mp. 125-131° C.
  • EXAMPLE 226
  • Figure US20060241123A1-20061026-C00252

    white solid
    Mp. 154-161° C.
  • EXAMPLE 227
  • Figure US20060241123A1-20061026-C00253

    white solid
    Mp. 179-184° C.
  • EXAMPLE 228
  • Figure US20060241123A1-20061026-C00254

    white solid
    Mp. 178-186° C.
  • EXAMPLE 229
  • Figure US20060241123A1-20061026-C00255

    colorless glassy solid
    Mp. 82-87° C.
  • EXAMPLE 230
  • Figure US20060241123A1-20061026-C00256

    colorless glassy solid
    Mp. 80-82° C.
  • EXAMPLE 231
  • Figure US20060241123A1-20061026-C00257

    white solid
    Mp. 117-125° C.
  • EXAMPLE 234
  • Figure US20060241123A1-20061026-C00258

    white solid
    Mp. 118-127° C.
    Preparation of the Carboxylic Acids:
    Synthesis Scheme for Carboxylic Acid 1
    Carboxylic Acid 1:
    Figure US20060241123A1-20061026-C00259
  • Intermediate CA1c) (0.1 g) was hydrolyzed by heating with hydrobromic acid (2 ml) and acetic acid (1.5 ml) for 3 h to give the desired compound after evaporation of the solvent.
    Intermediate CA1a):
    Figure US20060241123A1-20061026-C00260
  • To a solution of 2′-aminoacetophenone (709 mg) in methanol (10 ml) was added propionaldehyde (580 μl) and TMOF (482 μl) and the solution was stirred overnight. The volatiles were removed under reduced pressure and the residue was redissolved in methanol (10 ml). Acetic acid (115 μl) and sodium cyanoborohydride (126 mg) were added and the reaction was stirred overnight. After basification with 1 M NaOH the volatiles were removed under reduced pressure. The residue was dissolved in ethyl acetate and washed with water and brine. The organic phase was dried over Na2SO4 and the solvent was removed under reduced pressure to yield the title compound.
    Intermediate CA1b):
    Figure US20060241123A1-20061026-C00261
  • To a solution of intermediate CA1a) (681 mg) in dry THF (10 ml) was added TEA (670 μl) and the mixture was cooled to 0° C. At this temperature, ethyl oxalyl chloride (470 μl) was added dropwise. The reaction mixture was stirred for 4 h at room temperature. The volatiles were removed under reduced pressure and the residue was dissolved in ethyl acetate. The solution was washed with water and sat. NaHCO3 and brine and dried over Na2SO4 to yield the desired compound.
    Intermediate CA1c):
    Figure US20060241123A1-20061026-C00262
  • Intermediate CA1b) (555 mg) was dissolved in ethanol (10 ml). K2CO3 (276 mg) was added and the reaction mixture was stirred overnight. The reaction mixture was filtrated and the solvent was removed to yield the title compound.
    Figure US20060241123A1-20061026-C00263
  • Synthesis of Carboxylic Acids Using Method 1
  • Carboxylic Acid 2:
    Figure US20060241123A1-20061026-C00264
  • Intermediate CA2b) (5.85 g) was suspended in AcCl (110 ml) and concentrated sulfuric acid (4.40 ml) was added while stirring at RT. Then the slightly yellowish reaction mixture was heated to reflux with vigorous stirring and kept under reflux for 30 min. The reaction mixture was evaporated in vacuo to a volume of ca. 25 ml and then slowly and carefully added to well stirred H2O (300 ml) and stirring was continued for 1 h. After brief sonication, the formed precipitate was filtered off, washed with cold H2O (3×30 ml), and finally dried in vacuo at 40° C. overnight. The crude product was dissolved in a minimal amount of boiling H2O (270 ml) and left to slowly cool to RT. Crystallization was completed at RT for 6 h, then the crystalline product was filtered off and washed with cold H2O (3×10 ml). Finally the product was dried in vacuo at 40° C. overnight to yield the title compound.
    Intermediate CA2a):
    Figure US20060241123A1-20061026-C00265
  • 4-Trifluoromethoxyphenol (6.67 g) was dissolved in Et2O (55 ml) and TEA (6.36 ml) was added while stirring at RT. Then dimethyl acetylendicarboxylate (5.12 ml) was added with vigorous stirring and the reaction mixture stirred at RT in the dark overnight. The reaction mixture was diluted with Et2O (30 ml) and washed with 1 M HCl (3×65 ml), H2O (30 ml), and brine (25 ml), dried with Na2SO4 and then evaporated in vacuo. Finally it was dried under high vacuum for 2 h to yield the desired product.
    Intermediate CA2b):
    Figure US20060241123A1-20061026-C00266
  • To intermediate CA2a) (9.57 g) was added a solution of NaOH (4.80 g) in water (45 ml) while stirring at RT. Then the reaction mixture was heated to reflux with vigorous stirring and kept under reflux for 3 h. The reaction mixture was extracted with Et2O (100 ml) and then acidified to below pH 1 with conc. HCl while cooling in ice/H2O. A white precipitate formed, which was filtered off, washed with H2O (3×30 ml), and finally it was dried in vacuo at 40° C. overnight to give the desired compound.
  • The following chromone-2-carboxylic acids were prepared using method 1:
  • 6-ethylchromone-2-carboxylic acid, 6-isopropylchromone-2-carboxylic acid, 6-methoxychromone-2-carboxylic acid, 6-trifluoromethylchromone-2-carboxylic acid, 6-tert.-butylchromone-2-carboxylic acid, 6-chlorochromone-2-carboxylic acid, 6-trifluoromethoxychromone-2-carboxylic acid, 8-methoxychromone-2-carboxylic acid, 6-trifluoromethylsulfanylchromone-2-carboxylic acid, 8-chlorochromone-2-carboxylic acid, 8-fluorochromone-2-carboxylic acid 7-chlorochromone-2-carboxylic acid, 6-ethoxychromone-2-carboxylic acid, 6-methanesulfonylchromone-2-carboxylic acid, 8-oxo-8H-[1,3]dioxolo[4,5-g]chromene-6-carboxylic acid, 6-allyloxy-4-hydroxy-4H-chromene-2-carboxylic acid, 6-butoxy-4-hydroxy-4H-chromene-2-carboxylic acid, 6-propoxy-4-hydroxy-4H-chromene-2-carboxylic acid, 6-cyclopentyl-4-oxo-4H-chromene-2-carboxylic acid, 6-pentafluoroethoxy-4-oxo-4H-chromene-2-carboxylic acid, 4-oxo-6-[1,2,4]triazol-1-yl-4H-chromene-2-carboxylic acid, 6-imidazol-1-yl-4-oxo-4H-chromene-2-carboxylic acid, 6-acetylamino-4-oxo-4H-chromene-2-carboxylic acid, 6-(acetyl-methyl-amino)-4-oxo-4H-chromene-2-carboxylic acid, 6-methanesulfonylamino-4-oxo-4H-chromene-2-carboxylic acid, 6-(methanesulfonyl-methyl-amino)-4-oxo-4H-chromene-2-carboxylic acid and 6-dimethylamino-4-oxo-4H-chromene-2-carboxylic acid.
    Figure US20060241123A1-20061026-C00267
  • Synthesis of Carboxylic Acids Using Method 2
  • Carboxylic Acid 3:
    Figure US20060241123A1-20061026-C00268
  • Intermediate CA3a) (2.65 g) was suspended in sat. sodium bicarbonate solution (50 ml) and heated to 80° C. for 2 h. At the end of the reaction a clear solution was obtained. After cooling to room temperature the reaction mixture was acidified with HCl. The white precipitate was filtered off, washed with water and dried in vacuo at 40° C. overnight to give the title compound.
    Intermediate CA3a):
    Figure US20060241123A1-20061026-C00269
  • Sodium (4.0 g) was added to dry methanol (50 ml). After the conversion to the methoxide was complete the solution was cooled and a solution of 2′-hydroxy-4′,5′-dimethoxyacetophenone (3.92 g) in diethyl oxalate (12 ml), methanol (50 ml) and toluene (50 ml) was added to it. The mixture was refluxed overnight. After cooling, diethyl ether (200 ml) was added. The sodium salt was filtered, washed with anhydrous ether, suspended in water and the solution acidified. The resultant precipitate was filtered and dried to yield a yellow solid.
  • The intermediate was dissolved in ethanol (100 ml) and heated at 100° C. for 15 min; concentrated HCl (2 ml) was added, and the solution stirred at 100° C. for 1.5 h. Immediately after addition of the acid a precipitate was formed. After cooling to room temperature the reaction mixture was diluted with water (150 ml) and the pale yellow precipitate was filtered off and washed with water. The product was dried under reduced pressure.
  • The following chromone-2-carboxylic acids were prepared using method 2: 6-methoxychromone-2-carboxylic acid, 7-methoxychromone-2-carboxylic acid, 6,7-dimethylchromone-2-carboxylic acid, 6,7-dimethoxychromone-2-carboxylic acid, 6-chlorochromone-2-carboxylic acid, 6,8-difluorochromone-2-carboxylic acid, 6,8-dichlorochromone-2-carboxylic acid and 7-fluorochromone-2-carboxylic acid.
    Figure US20060241123A1-20061026-C00270
  • Demethylation of Methoxy Substituted Chromone-2-carboxylic Acids
  • Carboxylic Acid 4:
    Figure US20060241123A1-20061026-C00271
  • 8-Methoxychromone-2-carboxylic acid (220 mg) was suspended in AcOH (2 ml) and conc. HI (2 ml) was added while stirring at RT. Then the slightly yellowish suspension was heated to 120° C. with vigorous stirring and kept at this temperature for 60 min. The warm reaction mixture was slowly and carefully added to well stirred H2O (75 ml) and the resulting yellow solution was chilled in ice for 30 min. Crystallization was completed in the fridge for another 2 h. The formed crystalline precipitate was filtered off, washed with cold H2O (3×3 ml), and finally dried in vacuo at 40° C. overnight.
  • The following chromone-2-carboxylic acids were prepared using the demethylation method: 6-hydroxychromone-2-carboxylic acid, 7-hydroxychromone-2-carboxylic acid, 8-hydroxychromone-2-carboxylic acid, 6,7-dihydroxychromone-2-carboxylic acid and 6-hydroxy-7-methoxychromone-2-carboxylic acid.
  • Biological Assays
  • A. Binding Assay
  • A membrane binding assay is used to identify competitive inhibitors of fluorescence labeled NDP-alpha-MSH binding to HEK293 cell membrane preparations expressing human melanocortin receptors.
  • The test compound or unlabeled NDP-alpha-MSH is dispensed at varying concentrations to a 384 well microtiter plate. Fluorescence labeled NDP-alpha-MSH is dispensed at a single concentration, followed by addition of membrane preparations. The plate is incubated for 5 h at room temperature.
  • The degree of fluorescence polarization is determined with a fluorescence polarization microplate reader.
  • B. Functional Assay
  • A functional cellular assay, based on competition between unlabeled CAMP and a fixed quantity of fluorescence labeled cAMP for a limited number of binding sites on a cAMP specific antibody, is used to discriminate melanocortin receptor agonists from antagonists by fluorescence polarization.
  • HEK293 cells expressing one of the human melanocortin receptors are transferred to 384 well microtiter plates, an appropriate amount of cAMP antibody is added, followed by the addition of different concentrations of the test compound to effect cAMP production. Cells are lysed and a fluorescence labeled cAMP conjugate is dispensed. The plate is read on a fluorescence polarization microplate reader and the amount of cAMP produced as a response to a test compound is compared to the production of cAMP resulting from stimulation with NDP-alpha-MSH.
  • To define antagonistic activity of a test compound, the compound is dispensed at different concentrations to cells stimulated by an appropriate amount of NDP-α-MSH. Inhibition of cAMP production is determined by comparing the inhibition of cAMP production of the test compound to the inhibition of cAMP production by a known inhibitor tested at the same concentrations.
  • Biological Data for Selected Examples of the Invention:
    hMC4-R hMC4-R
    binding assay functional % activation
    Example IC50/nM assay EC50/nM functional assay
    1 80 no activation
    8 300 no activation
    15 4.4 700 40
    66 6.8 no activation
    110 790 no activation

    C. In Vivo Food Intake Models
    1. Spontaneous Feeding Paradigm
  • Food intake in rats is measured after i.p. or p.o. administration of the test compound (see e.g. Chen, A. S. et al. Transgenic Res 2000 April; 9(2):145-54).
  • Selected Examples of the present invention were active in the rat model at 3, 10 or 30 mg/kg after i.p. and p.o. administration of the test compound, respectively, using male Wistar rats (n=4−6).
  • Example 15 at 3 mg/kg p.o. administration lead to an increase in cumulative food intake of 290% (4 hours following administration p=0.048, n=4), 186% (6 hours following administration, p=0.108, n=4) and 355% (7 hours following administration, p=0.008, n=4), respectively, compared to control male Wistar rats receiving vehicle only (n=6).
  • Example 66 at 3 mg/kg p.o. administration lead to an increase in cumulative food intake of 410% (2 hours following administration p=0.057, n=4), 500% (4 hours following administration p=0.022, n=4), 186% (6 hours following administration, p=0.11, n=4) and 272% (7 hours following administration, p=0.02, n=4), respectively, compared to control male Wister rats receiving vehicle only (n=4).
  • Example 113 at 30 mg/kg p.o. administration lead to an increase in cumulative food intake of 192% (2 hours following administration p=0.083, n=4), 159% (4 hours following administration p=0.097, n=4), 191% (6 hours following administration, p=0.026, n=4) and 200% (7 hours following administration, p=0.013, n=4), respectively, compared to control male Wistar rats receiving vehicle only (n=8).
  • Example 212 at 10 mg/kg i.p. administration lead to an increase in cumulative food intake of 708% (2 hours following administration p=0.011, n=6), 725% (4 hours following administration p=0.009, n=6), 224% (6 hours following administration, p=0.046, n=6) and 167% (8 hours following administration, p=0.036, n=6), respectively, compared to control male Wistar rats receiving vehicle only (n=10).
  • 2. Model of LPS and Tumor-Induced Cachexia
  • Prevention or amelioration of cachexia induced by either lipopolysaccharide (LPS) administration or by tumor growth is determined upon i.p. or p.o. administration of test compounds to rats (see e.g. Marks, D. L.; Ling, N and Cone, R. D. Cancer Res 2001 Feb. 15; 61 (4): 1432-8).
  • A) Lipopolysaccharide-Induced Cachexia in Rats (Acute Model)
  • 1-2 Hours prior to the onset of the dark-phase, individually housed, male Wistar rats (200-300 g) receive an ip or po application of test-compound or vehicle (2 ml/kg, 1-30 mg/kg) which is followed or preceded by an ip injection of either lipopolysaccharide (LPS) or saline (2 ml/kg, 100 μg/kg). Food intake, water intake and body weight are measured at 1-24 hour intervals and differences between experimental groups are evaluated.
  • B) Tumour-Induced Cachexia in Mice (Chronic Model)
  • Subcutaneous injection of Lewis lung carcinoma cells to male C57BL6 mice (1 million cells/100 μl/mouse) results in non-metastasizing tumor growth which in turn results in loss of lean body mass. Chronic ip or po applications of test compounds (10 ml/kg, 1-30 mg/kg for 7-21 days) are accompanied by daily measurements of food intake, water intake and body weight. Lean body mass is measured at the start, during and at the termination of the study using magnetic resonance relaxometry, and at the end of the study using a conventional chemical extraction procedure (Soxhlet's extraction). Differences between experimental groups are evaluated.
  • Selected Examples of the present invention were active in the rat model at 10 mg/kg after p.o. administration of the test compound using male Wistar rats (n=5-6).
  • Example 66 at 10 mg/kg p.o. administration (in combination with LPS) lead to an increase in cumulative food intake of 148% (4 hours following administration p=0.04, n=5) compared to control male Wistar rats receiving LPS and vehicle only (n=6).
  • D. Rat Ex Copula Assay
  • Sexually mature male Caesarian Derived Sprague Dawley (CD) rats (over 60 days old) are used with the suspensory ligament surgically removed to prevent retraction of the penis back into the penile sheath during the ex copula evaluations. Animals receive food and water ad lib and are kept on a normal light/dark cycle. Studies are conducted during the light cycle.
  • 1. Conditioning to Supine Restraint for Ex Copula Reflex Tests
  • This conditioning takes about 4 days. Day 1, the animals are placed in a darkened restrainer and left for 15-30 minutes. Day 2, the animals are restrained in a supine position in the restrainer for 15-30 minutes. Day 3, the animals are restrained in the supine position with the penile sheath retracted for 15-30 minutes. Day 4, the animals are restrained in the supine position with the penile sheath retracted until penile responses are observed. Some animals require additional days of conditioning before they are completely acclimated to the procedures; non-responders are removed from further evaluation. After any handling or evaluation, animals are given a treat to ensure positive reinforcement.
  • 2. Ex Copula Reflex Tests
  • Rats are gently restrained in a supine position with their anterior torso placed inside a cylinder of adequate size to allow for normal head and paw grooming. For a 400-500 gram rat, the diameter of the cylinder is approximately 8 cm. The lower torso and hind limbs are restrained with a nonadhesive material (vetrap). An additional piece of vetrap with a hole in it, through which the glans penis will be passed, is fastened over the animal to maintain the preputial sheath in a retracted position. Penile responses will be observed, typically termed ex copula genital reflex tests. Typically, a series of penile erections will occur spontaneously within a few minutes after sheath retraction. The types of normal reflexogenic erectile responses include elongation, engorgement, cup and flip. An elongation is classified as an extension of the penile body. Engorgement is a dilation of the glans penis. A cup is defined as an intense erection where the distal margin of the glans penis momentarily flares open to form a cup. A flip is a dorsiflexion of the penile body.
  • Baseline and or vehicle evaluations are conducted to determine how, and if, an animal will respond. Some animals have a long duration until the first response while others are non-responders altogether. During this baseline evaluation latency to first response, number and type of responses are recorded. The testing time frame is 15 minutes after the first response.
  • After a minimum of 1 day between evaluations, these same animals are administered the test compound at 20 mg/kg and evaluated for penile reflexes. All evaluations are videotaped and scored later. Data are collected and analyzed using paired 2 tailed t-tests to compared baseline and/or vehicle evaluations to drug treated evaluations for individual animals. Groups of a minimum of 4 animals are utilized to reduce variability.
  • Positive reference controls are included in each study to assure the validity of the study. Animals can be dosed by a number of routes of administration depending on the nature of the study to be performed. The routes of administration includes intravenous (IV), intraperitoneal (IP), subcutaneous (SC) and intracerebral ventricular (ICV).
  • E. Models of Female Sexual Dysfunction
  • Rodent assays relevant to female sexual receptivity include the behavioral model of lordosis and direct observations of copulatory activity. There is also a urethrogenital reflex model in anesthetized spinally transected rats for measuring orgasm in both male and female rats. These and other established animal models of female sexual dysfunction are described in McKenna K E et al, A Model For The Study of Sexual Function In Anesthetized Male And Female Rats, Am. J. Physiol. (Regulatory Integrative Comp. Physiol 30): R1276-R1285, 1991; McKenna K E et al, Modulation By Peripheral Serotonin of The Threshold For Sexual Reflexes In Female Rats, Pharm. Bioch. Behav., 40:151-156, 1991; and Takahashi L K et al, Dual Estradiol Action In The Diencephalon And The Regulation of Sociosexual Behavior In Female Golden Hamsters, Brain Res., 359:194-207, 1985.
  • As evident from the results presented above, representative compounds of the present invention bind to the human melanocortin-4 receptor. Representative compounds of the present invention were also tested in the functional assay and found to be non-activating or activating the melanocortin-4 receptor with high EC50 values and low stimulation.
  • For example the compound consisting of the spiroindane “A moiety”, D-p-chlorophenylalanine and chromone-2-carboxylic acid as “C moiety”, example 1 of the present invention, binds to the melanocortin-4 receptor with an IC50 value of 80 nM and does not activate the melanocortin-4 receptor in the functional assay, whereas the compound with D-Tic as “C moiety” (WO9964002, example 13) is described to bind to the melanocortin-4 receptor with Ki=10 nM (Current Topics Med. Chem. 2003, 3, 855-883) and to activate the melanocortin-4 receptor with an EC50=190 nM (100% stimulation) (Bioorg. Med. Chem. Lett. 2003, 13, 3793-3796). Using our assay system the compound binds to the melanocortin-4 receptor with an IC50=54 nM. The receptor is activated with an EC50=1.1 μM (110% activation). Therefore example 1 is an antagonist whereas the compound with D-Tic is an agonist.
  • Compounds bearing an o-substituted arylpiperazine “A moiety” in combination with D-p-chlorophenylalanine as “B moiety” and D-Tic as “C moiety” are known to bind to the melanocortin-4 receptor with Ki values between 24 nM and 6.6 μM (J. Med. Chem. 2004, 47, 744-755, 29 examples) and to activate melanocortin-4 receptor in the functional assay with EC50 values between 14 nM and 1.3 μM (J. Med. Chem. 2004, 47, 744-755, 29 examples) and between 4 nM and 4.4 μM (Bioorg. Med. Chem. Lett. 2003, 13, 3793-3796, 23 examples). In the latter case three additional examples are reported to be weak agonists with 7- to 30-fold stimulation at 30 μM. There is no compound described which does not activate the melanocortin-4 receptor. Some of the compounds described above are claimed in patent applications WO03009850 (example 1) and WO02059108 (examples 2, 11, 15, 21, 24, 25, 26, 35, 37, 38, 40, 43, 44, 46, 52, 54, 56, 61, 74, 75, 79, 81, 153 and 154).
  • There is evidence in the literature that the stereochemistry of Tic in the “C moiety” does not have a big influence on the melanocortin-4 receptor affinity and activation at a concentration of 10 μM (J. Med. Chem. 2002, 45, 4589-4593, compounds 1 and 13). Both diastereomers activate the melanocortin-4 receptor, however, the D-Tic derived compound 1 shows improved functional activity. Applying this concept to compounds bearing the o-substituted arylpiperazine “A moiety” it can be concluded that the diastereomers of the compounds described in the literature cited above also act as agonists. Some of the L-Tic derivatives are described in WO03009850 (examples 56, 66, 91, 93, 117, 118 and 423).
  • Example 8 of the present invention binds to the melanocortin-4 receptor with an IC50=300 nM and does not activate the melanocortin-4 receptor in the functional assay. The corresponding compound with D-Tic as “C moiety” (WO03009850, example 1 and WO02059108, example 154) is described to bind to the melanocortin-4 receptor with Ki=220 nM and to activate said receptor with an EC50=16 nM (J. Med. Chem. 2004, 47, 744-755, compound 39). Another source describes said compound to activate the melanocortin-4 receptor with an EC50=380 nM (100% stimulation) (Bioorg. Med. Chem. Lett. 2003, 13, 3793-3796, compound 3). In our assays the compound was found to have a melanocortin-4 receptor binding IC50=500 nM and to activate the melanocortin-4 receptor with an EC50=3.7 μM (96% activation). Therefore example 8 is an antagonist whereas the compound with D-Tic is an agonist.
  • Example 110 of the present invention binds to the melanocortin-4 receptor with an IC50=790 nM and does not activate the melanocortin-4 receptor in the functional assay. The corresponding compound with D-Tic as “C moiety” (WO02059108, example 56) is described to bind to the melanocortin-4 receptor with Ki=210 nM and to activate said receptor with an EC50=48 nM (J. Med. Chem. 2004, 47, 744-755, compound 43). Therefore example 110 is an antagonist whereas the compound with D-Tic is an agonist.
  • Compounds having a 4,4-disubstituted piperidine “A moiety” were generally found to be more potent ligands for the melanocortin-4 receptor.
  • Example 15 bearing the 4-cyclohexyl-4-[1,2,4]triazol-1-ylmethyl-piperidine “A moiety” binds to the melanocortin-4 receptor with an IC50=4.4 nM. In the functional assay the compound acts as a partial agonist of said receptor with an EC50=700 nM (40% activation). The corresponding compound with D-Tic as “C moiety” (WO0074679, example 2) is described to bind to the melanocortin-4 receptor with an IC50 between 0.92 and 7 nM and to act as a full agonist of the melanocortin-4 receptor with an EC50 between 2.1 and 4 nM (WO0074679, example 2; J. Med. Chem. 2002, 45, 4589-4593, compound 1; Bioorg. Med. Chem. Lett. 2003, 13, 4341-4344, compound 1 and Bioorg. Med. Chem. Lett. 2003, 13, 3793-3796, compound 27). In our assay system the compound binds to the melanocortin-4 receptor with an IC50=10 nM and activates the receptor with an EC50=50 nM (102% activation) when tested in the functional assay. Therefore example 15 is a very weak agonist whereas the compound with D-Tic is a full agonist.
  • Example 66 having the 4-cyclohexyl-piperidine-4-carboxylic acid tert-butylamide “A moiety” has a similar affinity to the melanocortin-4 receptor than example 15. The compound binds to the receptor with IC50=6.8 nM, however, no activation of said receptor is observerd in the functional assay. Therefore example 66 is an antagonist.
  • As illustrated by the biological results (see above) representative compounds of the present invention are also active as antagonists when tested in vivo.
  • Example 15 is active in the spontaneous feeding paradigm at a dose of 3 mg/kg when administered orally. The test animals show a significantly increase in food intake. However, the corresponding compound with D-Tic as “C moiety” is reported to act as in vivo agonist in a conscious rat model of reflexogenic erections when administered i.v. or p.o. (WO0074679, example 2) and it was also evaluated for its effects oh food intake (J. Med. Chem. 2002, 45, 4589-4593, compound 1). Food intake was significantly reduced on administration of the compound.
  • Example 66 is active in the spontaneous feeding paradigm as well as in the model of LPS-induced cachexia. In the spontaneous feeding paradigm the test animals show a significant increase in food intake at dose of 3 mg/kg p.o. and in the acute model of lipopolysaccharide-induced cachexia the test animals show a significant increase in cumulative food intake at a dose of 10 mg/kg p.o.
  • Examples of a Pharmaceutical Composition
  • As a specific embodiment of an oral composition of a compound of the present invention, 25 mg of Example 66 is formulated with sufficient finely divided lactose to provide a total amount of 580 to 590 mg to fill a size 0 hard gelatin capsule.
  • As another specific embodiment of an oral composition of a compound of the present invention, 35 mg of Example 113 is formulated with sufficient finely divided lactose to provide a total amount of 580 to 590 mg to fill a size 0 hard gelatin capsule.
  • While the invention has been described and illustrated in reference to certain preferred embodiments thereof, those skilled in the art will appreciate that various changes, modifications and substitutions can be made therein without departing from the spirit and scope of the invention. For example, effective dosages, other than the preferred doses as set forth above, may be applicable as a consequence of the specific pharmacological responses observed and may vary depending upon the particular active compound selected, as well as from the type of formulation and mode of administration employed, and such expected variations or differences in the results are contemplated in accordance with the objects and practices of the present invention. It is intended, Thereforee, that the invention be limited only by the scope of the claims which follow and that such claims be interpreted as broadly as is reasonable.

Claims (14)

1. A compound of structural formula (I):
Figure US20060241123A1-20061026-C00272
or a pharmceutically acceptable salt or a solvate thereof, wherein
Ar is:
aryl or heteroaryl which may both be substituted;
R1 is:
Figure US20060241123A1-20061026-C00273
A is:
Figure US20060241123A1-20061026-C00274
R2 is independently:
hydrogen,
halo,
alkyl,
haloalkyl,
hydroxy,
alkoxy,
S-alkyl,
SO2-alkyl,
O-alkenyl,
S-alkenyl,
NR14C(O)R14,
NR14SO2R14,
N(R14)2,
(D)-cycloalkyl,
(D)-aryl,
(D)-heteroaryl,
(D)-heterocyclyl (wherein heterocyclyl excludes a heterocyclyl containing a single nitrogen), and
wherein aryl, heteroaryl, heterocyclyl, alkyl and cycloalkyl are substituted or unsubstituted, and two adjacent R2 may form a 4- to 7-membered ring;
R4 and R5 are each independently:
hydrogen, alkyl or
(D)-cycloalkyl, or
R4 and R5 together with the nitrogen to which they are attached form a 5- to 8-membered ring,
wherein alkyl and cycloalkyl are unsubstituted or substituted;
R8 is independently:
hydrogen,
alkyl,
(D)-aryl or
(D)-cycloalkyl;
R9 is independently:
hydrogen,
alkyl,
(D)-aryl,
(D)-heteroaryl or
(D)-cycloalkyl;
R10 is independently:
R9,
(D)-heterocyclyl,
(D)-N(Y)2,
(D)-NH-heteroaryl or
(D)-NH-heterocyclyl,
wherein aryl, heteroaryl, alkyl, D, cycloalkyl and heterocyclyl are substituted or unsubstituted, or
two R10 groups together with the atoms to which they are attached form a 5- to 8-membered mono- or bi-cyclic ring system;
R11 is:
hydrogen,
halo,
alkyl,
alkoxy,
C≡N,
CF3 or
OCF3;
R12 is independently:
hydrogen,
hydroxy,
cyano,
nitro,
halo,
alkyl,
alkoxy,
haloalkyl,
(D)-C(O)R14,
(D)-C(O)OR14,
(D)-C(O)SR14,
(D)-C(O)-heteroaryl,
(D)-C(O)-heterocyclyl,
(D)-C(O)N(R14)2,
(D)-N(R14)2,
(D)-NR14COR14,
(D)-NR14CON(R14)2,
(D)-NR14C(O)OR14,
(D)-NR14C(R14)═N(R14),
(D)-NR14C(═NR14)N(R14)2,
(D)-NR14SO2R14,
(D)-NR14SO2N(R14)2,
(D)-NR14(D)-heterocyclyl,
(D)-NR14(D)-heteroaryl,
(D)-OR14,
OSO2R14,
(D)-[O]q(cycloalkyl),
(D)-[O]q(D)aryl,
(D)-[O]q(D)-heteroaryl,
(D)-[O]q(D)-heterocyclyl (wherein heterocyclyl excludes a heterocyclyl containing a single nitrogen when q=1),
(D)-SR14,
(D)-SOR14,
(D)-SO2R14 or
(D)-SO2N(R14),
wherein alkyl, alkoxy, cycloalkyl, aryl, heterocyclyl and heteroaryl are substituted or unsubstituted;
R14 is independently:
hydrogen,
alkyl,
haloalkyl,
(D)-cycloalkyl,
(D)-phenyl,
(D)-naphthyl,
(D)-heteroaryl,
(D)-heterocyclyl (wherein heterocyclyl excludes a heterocyclyl containing a single nitrogen), and
wherein phenyl, naphthyl, heteroaryl, heterocyclyl, alkyl and cycloalkyl are substituted or unsubstituted;
X is:
alkyl,
(D)-cycloalkyl,
(D)-aryl,
(D)-heteroaryl,
(D)-heterocyclyl,
(D)-C≡N,
(D)-CON(R9R9),
(D)-CO2R9,
(D)-COR9,
(D)-NR9C(O)R9,
(D)-NR9CO2R9,
(D)-NR9C(O)N(R9)2,
(D)-NR9SO2R9,
(D)-S(O)pR9,
(D)-SO2N(R9)(R9),
(D)-OR9,
(D)-OC(O)R9,
(D)-OC(O)OR9,
(D)-OC(O)N(R9)2,
(D)-N(R9)(R9) or
(D)-NR9SO2N(R9)(R9),
wherein aryl, heteroaryl, alkyl, D, cycloalkyl and heterocyclyl are unsubstituted or substituted;
Y is:
hydrogen,
alkyl,
(D)-cycloalkyl,
(D)-aryl,
(D)-heterocyclyl or
(D)-heteroaryl,
wherein aryl, heteroaryl, alkyl, D and cycloalkyl are unsubstituted or
substituted;
Cy is benzene, pyridine or cyclohexane;
D is a bond or alkylene;
E is CHCO2Y, CHC(O)N(Y)2, NSO2R10, CHN(Y)COR10, CHN(Y)SO2R10, CHCH2OY or CHCH2heteroaryl;
G is D, CH-alkyl, O, C═O or SO2, with the proviso that when G is O, the ring atom E is carbon;
J is N or CH;
T is O;
n is 0-2;
m is 0-2;
o is 0-3;
p is 0-2;
q is 0 or 1;
r is 1 or 2.
2. The compound of claim 1, wherein
Ar is:
aryl which may be substituted with one to three substituents independently selected from the group consisting of cyano, nitro, perfluoroalkoxy, halo, alkyl, (D)-cycloalkyl, alkoxy and/or haloalkyl;
R1 is:
Figure US20060241123A1-20061026-C00275
A is:
Figure US20060241123A1-20061026-C00276
R2 is independently:
hydrogen,
hydroxy,
halo,
alkyl,
alkoxy,
S-alkyl,
SO2-alkyl,
O-alkenyl,
S-alkenyl,
haloalkyl or
(D)-cycloalkyl;
R4 and R5 are each independently:
hydrogen,
alkyl or
cycloalkyl, or
R4 and R5 together with the nitrogen to which they are attached form a 5- to 7-membered ring which may contain an additional heteroatom selected from O, S and NR6;
R6 is independently:
hydrogen,
alkyl,
C(O)alkyl,
(D)-aryl or
(D)-cycloalkyl;
R8 is independently:
hydrogen,
alkyl or
(D)-aryl;
R9 is independently:
hydrogen,
alkyl or
(D)-cycloalkyl;
R10 is R9;
R11 is:
hydrogen,
halo,
alkyl,
alkoxy or
C≡N;
R12 is independently:
hydrogen,
hydroxy,
cyano,
nitro,
halo,
alkyl,
alkoxy,
haloalkyl,
(D)-C(O)-heterocyclyl,
(D)-C(O)N(R14)2,
(D)-N(R14)2,
(D)-NR14COR14,
(D)-NR14CON(R14)2,
(D)-NR14C(O)OR14,
(D)-NR14C(R14)═N(R14),
(D)-NR14C(═NR14)N(R14)2,
(D)-NR14SO2R14 or
(D)-NR14SO2N(R14)2;
R14 is independently:
hydrogen,
halo,
alkyl,
(D)-cycloalkyl,
alkoxy or
phenyl;
X is:
alkyl,
(D)-cycloalkyl,
(D)-aryl,
(D)-heteroaryl,
(D)-heterocyclyl,
(D)-NHC(O)R9,
(D)-CO2R9 or
(D)-CON(R9R9);
Y is:
hydrogen,
alkyl,
(D)-cycloalkyl,
(D)-aryl,
(D)-heterocyclyl or
(D)-heteroaryl;
Cy is benzene or pyridine;
D is a bond or C1-C4-alkylene;
E is NSO2R10 CHN(Y)COR10 or CHN(Y)SO2R10;
G is D or CH-alkyl;
J is N or CH;
n is 0 or 1;
m is 0 or 1;
o is 0, 1 or 2;
r is 1.
3. The compound of claim 1, wherein
Ar is:
phenyl or naphthyl which may be substituted with one or two substituents independently selected from the group consisting of halo, alkyl, alkoxy and/or haloalkyl;
R1 is:
Figure US20060241123A1-20061026-C00277
A is:
Figure US20060241123A1-20061026-C00278
R2 is independently:
hydrogen,
hydroxy,
alkoxy,
S-alkyl,
SO2-alkyl,
O-alkenyl,
S-alkenyl,
halo or
alkyl;
R4 and R5 are each independently:
hydrogen or
alkyl, or
R4 and R5 together with the nitrogen to which they are attached form a 5- to 6-membered ring optionally containing an additional oxygen atom;
R6 is hydrogen;
R8 is independently:
alkyl or
(D)-aryl;
R9 is alkyl;
R10 is R9;
R11 is:
hydrogen,
halo and
C1-C4-alkyl;
R12 is independently:
cyano,
nitro,
halo,
alkyl,
(D)-C(O)-heterocyclyl,
(D)-N(R14)2,
(D)-NR14COR14,
(D)-NR14CON(R14)2,
(D)-NR14C(O)OR14 or
(D)-NR14SO2R14;
R14 is independently:
hydrogen,
halo,
alkyl,
alkoxy or
phenyl;
X is:
Alkyl,
(D)-cycloalkyl,
(D)-heterocyclyl,
(D)-NHC(O)R9 or
(D)-CON(R9R9);
Y is:
hydrogen,
alkyl,
(D)-cycloalkyl or
(D)-heterocyclyl;
Cy is benzene;
D is a bond or CH2;
E is NSO2R10;
G is D;
J is N or CH;
n is 0;
m is 0;
o is 0 or 1;
p is 0, 1 or 2;
q is 0 or 1;
r is 1.
4. A medicament comprising the compound of claim 1.
5. A method of treating or preventing disorders, diseases or conditions responsive to the modulation of the melanocortin-4 receptor in a mammal, where modulation means activation in the case of MC-4-R agonists or inactivation in the case of MC-4-R antagonists, the method comprising administering an effective amount of a compound of claim 1.
6. A method of treating or preventing cancer cachexia, the method comprising administering to a human or mammal an effective amount of the MC-4-R antagonists according to claim 5.
7. A method of treating or preventing muscle wasting, the method comprising administering to a human or mammal an effective amount of the MC-4-R antagonists according to claim 5.
8. A method of treating or preventing anorexia, the method comprising administering to a human or mammal an effective amount of the MC-4-R antagonists according to claim 5.
9. A method of treating or preventing anxiety and/or depression, the method comprising administering to a human or mammal an effective amount of the MC-4-R antagonists according to claim 5.
10. A method of treating or preventing obesity, the method comprising administering to a human or mammal an effective amount of the MC-4-R antagonists according to claim 5.
11. A method of treating or preventing diabetes mellitus, the method comprising administering to a human or mammal an effective amount of the MC-4-R antagonists according to claim 5.
12. A method of treating or preventing male or female sexual dysfunction, the method comprising administering to a human or mammal an effective amount of the MC-4-R antagonists according to claim 5.
13. A method of treating or preventing erectile dysfunction, the method comprising administering to a human or mammal an effective amount of the MC-4-R antagonists according to claim 5.
14. A pharmaceutical composition which comprises a compound of claim 1 and a pharmaceutically acceptable carrier.
US10/550,267 2003-03-20 2004-03-19 Substituted piperidine and piperazine derivatives as melanocortin-4 receptor modulators Abandoned US20060241123A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP03006254.1 2003-03-20
EP03006254A EP1460073A1 (en) 2003-03-20 2003-03-20 Substituted piperidine and piperazine derivatives as melanocortin-4 receptor modulators
PCT/EP2004/002907 WO2004083209A1 (en) 2003-03-20 2004-03-19 Substituted piperidine and piperazine derivatives as melanocortin-4 receptor modulators

Publications (1)

Publication Number Publication Date
US20060241123A1 true US20060241123A1 (en) 2006-10-26

Family

ID=32798840

Family Applications (1)

Application Number Title Priority Date Filing Date
US10/550,267 Abandoned US20060241123A1 (en) 2003-03-20 2004-03-19 Substituted piperidine and piperazine derivatives as melanocortin-4 receptor modulators

Country Status (8)

Country Link
US (1) US20060241123A1 (en)
EP (2) EP1460073A1 (en)
JP (1) JP2006520362A (en)
AT (1) ATE367391T1 (en)
AU (1) AU2004222095B2 (en)
CA (1) CA2519442A1 (en)
DE (1) DE602004007633D1 (en)
WO (1) WO2004083209A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2520114A1 (en) 2003-04-04 2004-10-21 Merck & Co., Inc. Acylated spiropiperidine derivatives as melanocortin-4 receptor agonists
WO2021133563A1 (en) * 2019-12-23 2021-07-01 Crinetics Pharmaceuticals, Inc. Spirocyclic piperidine melanocortin subtype-2 receptor (mc2r) antagonists and uses thereof
CN116113635A (en) 2020-06-09 2023-05-12 辉瑞公司 Spiro compounds as melanocortin 4 receptor antagonists and uses thereof

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2166975C (en) * 1993-07-16 2005-04-05 Mark G. Bock Benzoxazinone and benzopyrimidinone piperidinyl tocolytic oxytocin receptor antagonists
US5968929A (en) * 1996-10-30 1999-10-19 Schering Corporation Piperazino derivatives as neurokinin antagonists
CA2334551A1 (en) * 1998-06-11 1999-12-16 Merck & Co., Inc. Spiropiperidine derivatives as melanocortin receptor agonists
AU766191B2 (en) * 1999-06-04 2003-10-09 Merck & Co., Inc. Substituted piperidines as melanocortin-4 receptor agonists
US6432980B1 (en) * 1999-11-12 2002-08-13 Merck & Co., Inc. Aliphatic amine substituted piperidyl diaryl pyrrole derivatives as antiprotozoal agents
US6472398B1 (en) * 2000-03-23 2002-10-29 Merck & Co., Inc. Spiropiperidine derivatives as melanocortin receptor agonists
AU2001264977B2 (en) * 2000-05-30 2005-04-14 Merck & Co., Inc. Melanocortin receptor agonists
JP2004532838A (en) * 2001-03-02 2004-10-28 ブリストル−マイヤーズ スクイブ カンパニー Compounds useful as melanocortin receptor modulators and pharmaceutical compositions containing them
US6977264B2 (en) * 2001-07-25 2005-12-20 Amgen Inc. Substituted piperidines and methods of use

Also Published As

Publication number Publication date
CA2519442A1 (en) 2004-09-30
AU2004222095A1 (en) 2004-09-30
EP1603912B1 (en) 2007-07-18
DE602004007633D1 (en) 2007-08-30
EP1603912A1 (en) 2005-12-14
EP1460073A1 (en) 2004-09-22
JP2006520362A (en) 2006-09-07
ATE367391T1 (en) 2007-08-15
AU2004222095B2 (en) 2007-05-17
WO2004083209A1 (en) 2004-09-30

Similar Documents

Publication Publication Date Title
US20070155783A1 (en) Substituted cyclohexyl and piperidinyl derivates as melanocortin-4 receptor modulators
EP2176250B1 (en) Substituted heteroarylpiperidine derivatives as melanocortin-4 receptor modulators
EP1842846A1 (en) Phenylpiperidine derivatives as melanocortin-4 receptor modulators
US20070037823A1 (en) Substituted piperidine and piperazine derivatives as melanocortin-4 receptor modulators
EP1538159A1 (en) Substituted N-benzyl-lactam derivatives as melanocortin-4 receptor agonists
WO2010081666A1 (en) Substituted heteroarylpiperidine derivatives as melanocortin-4 receptor modulators
US20060241123A1 (en) Substituted piperidine and piperazine derivatives as melanocortin-4 receptor modulators
US20060276485A1 (en) Substituted piperidine and piperazine derivatives as melanocortin-4 receptor modulators
EP2020405A1 (en) Substituted aryl or heteroarylpiperidine derivatives as melanocortin-4 receptor modulators
EP1826201A1 (en) Substituted phenylpiperidine derivatives as melanocortin-4 receptor modulators

Legal Events

Date Code Title Description
AS Assignment

Owner name: SANTHERA PHARMACEUTICALS (SCHWEIZ) GMBH, SWITZERLA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SOEBERDT, MICHAEL;WEYERMANN, PHILIPP;VON SPRECHER, ANDREAS;REEL/FRAME:017088/0333

Effective date: 20051024

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION